US10858450B2 - Bi-specific fusion proteins - Google Patents

Bi-specific fusion proteins Download PDF

Info

Publication number
US10858450B2
US10858450B2 US15/957,252 US201815957252A US10858450B2 US 10858450 B2 US10858450 B2 US 10858450B2 US 201815957252 A US201815957252 A US 201815957252A US 10858450 B2 US10858450 B2 US 10858450B2
Authority
US
United States
Prior art keywords
fusion protein
specific fusion
polypeptide
domain
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US15/957,252
Other versions
US20180237540A1 (en
Inventor
Ulrik Bjerl Nielsen
Thomas Wickham
Birgit Schoeberl
Brian Harms
Bryan Linggi
Matthew Onsum
Byron Delabarre
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Silver Creek Pharmaceuticals Inc
Original Assignee
Silver Creek Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Silver Creek Pharmaceuticals Inc filed Critical Silver Creek Pharmaceuticals Inc
Priority to US15/957,252 priority Critical patent/US10858450B2/en
Assigned to MERRIMACK PHARMACEUTICALS, INC reassignment MERRIMACK PHARMACEUTICALS, INC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LINGGI, BRYAN, DELABARRE, BYRON, ONSUM, MATTHEW, NIELSEN, ULRIK, WICKHAM, THOMAS, SCHOEBERL, BIRGIT, HARMS, BRIAN
Publication of US20180237540A1 publication Critical patent/US20180237540A1/en
Assigned to Silver Creek Pharmaceuticals, Inc. reassignment Silver Creek Pharmaceuticals, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MERRIMACK PHARMACEUTICALS, INC.
Priority to US17/078,978 priority patent/US11673970B2/en
Application granted granted Critical
Publication of US10858450B2 publication Critical patent/US10858450B2/en
Priority to US18/311,306 priority patent/US20230322951A1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/66Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid the modifying agent being a pre-targeting system involving a peptide or protein for targeting specific cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6891Pre-targeting systems involving an antibody for targeting specific cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/485Epidermal growth factor [EGF] (urogastrone)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/65Insulin-like growth factors (Somatomedins), e.g. IGF-1, IGF-2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/80Fusion polypeptide containing a DNA binding domain, e.g. Lacl or Tet-repressor

Definitions

  • the present invention relates generally to fusion proteins that have therapeutic uses, and more specifically to bi-specific fusion proteins, pharmaceutical compositions comprising such fusion proteins, and methods for using such fusion proteins to repair damaged tissue.
  • Myocardial infarction commonly known as a heart attack, occurs when coronary artery obstruction cuts off the blood supply to part of the heart.
  • the resulting lack of oxygen causes irreversible tissue damage (necrosis and apoptosis), due to the inability of the heart to sufficiently activate endogenous regeneration programs and self-repair.
  • tissue damage is a leading cause of congestive heart failure, a condition in which the heart is no longer capable of effectively pumping blood.
  • congestive heart failure In the United States, there are more than a million heart attacks every year, and nearly 5 million people are afflicted with congestive heart failure.
  • Stem cell therapy is a potential new strategy for cardiac repair.
  • it is possible to generate cardiac muscle cells from stem cells. This suggests that stems cells could be used to repair damaged tissue such as cardiac tissue in a patient; however, no therapeutic treatments based on such an approach are presently available.
  • One difficulty that has been encountered in stem cell therapy is that of targeting sufficient numbers of stem cells to the damaged tissue to result in clinically significant repair.
  • the present invention provides bi-specific fusion proteins, nucleic acid molecules encoding bi-specific fusion proteins and therapeutic methods that employ such bi-specific fusion proteins.
  • the present invention provides bi-specific fusion proteins that comprise: (a) a targeting domain having a binding specificity to an ischemia-associated molecule; and (b) an activator domain having a binding specificity to a growth factor receptor or cytokine receptor, wherein upon exposure of the activator domain to the growth factor receptor or cytokine receptor, the activator domain binds the growth factor receptor or cytokine receptor so as to modulate regeneration of a cardiac tissue.
  • the bi-specific protein comprises: (a) a targeting polypeptide domain that binds to an ischemia-associated molecule with a K d (i.e., said binding exhibits a K d ) ranging from 10 ⁇ 6 to 10 ⁇ 12 M or better; and (b) an activator domain that that detectably modulates the activity of a cellular network (e.g., detectably modulates activation of a growth factor receptor or cytokine receptor).
  • the targeting polypeptide domain binds to the ischemia-associated molecule with a K d ranging from 10 ⁇ 7 to 10 ⁇ 12 M or better, or ranging from 10 ⁇ 8 to 10 ⁇ 12 M or better.
  • the K d is determined using a biosensor, e.g., by surface plasmon resonance or resonant mirror analysis.
  • certain bi-specific fusion proteins further comprise: (c) a polypeptide linker wherein the polypeptide linker extends the half life of the bi-specific fusion protein.
  • the targeting domain is at the N-terminus or at the C-terminus of the activator domain.
  • the polypeptide linkers at the N-terminus or at the C-terminus of the targeting domain.
  • the targeting domain is at the amino terminus of the fusion protein and the activator domain is at the carboxy terminus of the fusion protein.
  • the targeting domain is at the carboxy terminus of the fusion protein and the activator domain is at the amino terminus of the fusion protein.
  • the polypeptide linker has two termini, an N-terminus and a C-terminus, that is joined at one terminus via a peptide bond to the targeting polypeptide domain and is joined at the other terminus via a peptide bond to the activator domain.
  • the targeting peptide is linked to the N-terminus of the linker and the activator domain is linked to the C-terminus of the linker.
  • the targeting peptide is linked to the C-terminus of the linker and the activator domain is linked to the N-terminus of the linker.
  • the linker is non-immunogenic in humans (e.g., a human serum protein or derivative thereof).
  • linkers comprise at least 100 consecutive amino acids that are at least 80% identical to a serum albumin amino acid sequence, such as a human alpha-fetoprotein sequence.
  • the linker comprises or has an amino acid sequence recited in any one of SEQ ID NOs: 10-29.
  • the bi-specific fusion protein comprises (a) a targeting domain having a binding specificity to an ischemia-associated molecule; (b) an activator domain that detectably modulates activation of a receptor; and (c) a polypeptide linker, wherein the polypeptide linker extends the half life of the bi-specific fusion protein.
  • the bi-specific fusion protein comprises (a) a targeting domain having a binding specificity to a target molecule; (b) an activator domain having a binding specificity to a receptor, wherein upon exposure of the activator domain to the receptor, the activator domain binds the receptor so as to modulate activation of the receptor; and (c) a polypeptide linker, wherein the polypeptide linker extends the half life of the bi-specific fusion protein.
  • the bi-specific protein comprises (a) a targeting domain having a binding specificity to a tissue-associated molecule; and (b) an activator domain having a binding specificity to a molecule associated with the surface of a cell in the tissue, wherein upon exposure of the activator domain to surface-associated molecule, the activator domain binds the membrane-associated molecule so as to modulate regeneration of the tissue, wherein the targeting domain and the activator domain are linked via a linker, and wherein the linker extends the half life of the bi-specific fusion protein.
  • the bi-specific fusion protein comprises (a) a targeting domain having a binding specificity to a target molecule; (b) an activator domain having a binding specificity to a receptor, wherein upon exposure of the activator domain to the receptor, the activator domain binds the receptor so as to modulate tissue regeneration; and (c) a polypeptide linker, wherein the polypeptide linker extends the half life of the bi-specific fusion protein.
  • the targeting domain binds to the molecule with a dissociation constant Kd ranging from 10 ⁇ 6 M to 10 ⁇ 12 M.
  • the targeting domain binds to a molecule selected from the group of mysosin, cardiac myosin, DNA, phosphatidylserine, collagen, or extracellular matrix proteins.
  • the targeting domain can be selected from the group of annexin, anti-myosin antibody, anti-DNA scFv, variants thereof, fragments thereof, and combinations thereof.
  • the scFv antibody has a sequence recited in any one of SEQ ID NOs: 1, 2, or 30.
  • annexin has a sequence recited in SEQ ID NO: 31.
  • the activator domain binds specifically to a growth factor receptor or cytokine receptor.
  • the activator domain is selected from the group consisting of hepatocyte growth factor, vascular endothelial growth factor, fibroblast growth factor, neuregulin/heregulin, variant thereof, and portion thereof.
  • the bi-specific fusion proteins comprises (a) a leader polypeptide that comprises a sequence recited in SEQ ID NO:41 or 42; (b) a targeting polypeptide domain that binds to an ischemia-associated molecule, said binding exhibiting a K d ranging from 10 ⁇ 6 to 10 ⁇ 12 M or better; (c) a short connector polypeptide that comprises the sequence -Gly-Ala- or -Ala-Ser-; (d) a HSA polypeptide that comprises a sequence recited in any one of SEQ ID NOs:10, 12, 14-29 and 45); (e) a short connector polypeptide that comprises the sequence -Leu-Gln- or -Thr-Gly-; (f) an activator domain that that detectably modulates the activity of a cellular network; and (g) a hexahistidine-comprising polypeptide.
  • the targeting polypeptide domain comprises a sequence recited in SEQ ID NO:1, 2, 30 or 31;
  • the HSA polypeptide comprises the sequence recited in SEQ ID NO:45;
  • the activator domain comprises a sequence recited in any one of SEQ ID NOs: 32-40; and
  • the hexahistidine-comprising polypeptide has a sequence recited in SEQ ID NO:43 or 44.
  • the ischemia-associated molecule is a DNA molecule, myosin (e.g., a myosin subtype such as cardiomyosin) or phosphatidyl serine.
  • the targeting polypeptide comprises an antibody variable region.
  • the targeting polypeptide comprises a scFv antibody.
  • Representative such scFv antibodies comprise or have a sequence recited in SEQ ID NO:1 or SEQ ID NO:2.
  • the activator domain is a growth factor polypeptide.
  • the growth factor polypeptide binds to a receptor for IGF or HGF (e.g., the growth factor polypeptide comprises or has an amino acid sequence recited in any one of SEQ ID NOs:3-9).
  • the bi-specific binding agents provided herein are not necessarily limited to two binding specificities.
  • the bi-specific fusion protein in addition to the targeting domain, comprises two or more activator domains that are linked directly or indirectly via peptide bonds and are selected from growth factor polypeptides and cytokine polypeptides.
  • the present invention provides pharmaceutical compositions, comprising a bi-specific fusion protein as described above in combination with a physiologically acceptable carrier.
  • methods for treating pathological tissue damage in a patient, comprising administering a pharmaceutical composition to a patient suffering from pathological tissue damage, and thereby decreasing pathological tissue damage in the patient.
  • the pathological tissue damage is heart tissue damage associated with myocardial infarction.
  • the pathological tissue damage is kidney tissue damage.
  • methods for promoting tissue regeneration in a patient.
  • the methods comprise (a) providing a bi-specific fusion protein comprising (i) a targeting domain having a binding specificity to an ischemia-associated molecule; and (ii) an activator domain having a binding specificity to growth factor receptor or cytokine receptor; and (b) administering in a patient in need thereof a therapeutically effective amount of the bi-specific fusion protein whereby the targeting domain specifically binds to the ischemia-associated molecule thereby targeting the bi-specific fusion protein to a tissue and whereby upon exposure of the activator domain to the growth factor receptor or cytokine receptor, the activator domain specifically activates the growth factor receptor or cytokine receptor so as to promote tissue regeneration.
  • the methods comprise (a) providing a bi-specific fusion protein comprising (i) a targeting domain having a binding specificity to a target molecule; (ii) an activator domain having a binding specificity to a receptor; (iii) a polypeptide linker, wherein the polypeptide linker extends the half life of the bi-specific fusion protein; and (b) administering in a patient in need thereof a therapeutically effective amount of the bi-specific fusion protein whereby the targeting domain specifically binds to the target molecule thereby targeting the bi-specific fusion protein to a first cell and whereby upon exposure of the activator domain to the growth factor receptor, the activator domain specifically activates the receptor of a second cell of a tissue so as to promote tissue regeneration.
  • such methods further comprise the administration of stem cells to the patient.
  • the bi-specific protein upon administration of the bi-specific protein, prevents cell damage, increases survival, promotes cell growth, promotes motility of stem cells, recruits stem cells, promotes differentiation of stem cells.
  • nucleic acid molecules encoding a bi-specific fusion protein as described above.
  • the nucleic acid molecule is DNA, and the DNA further comprises transcriptional and translational regulatory sequences operably linked to the bi-specific fusion protein coding sequence, such that transcription and translation of the coding sequence occurs in at least one eukaryotic cell type.
  • SEQ ID NO:1 is the amino acid sequence of the anti-DNA scFv SV-1.
  • SEQ ID NO:2 is the amino acid sequence of the anti-DNA scFv SV-22.
  • SEQ ID NO:3 is the amino acid sequence of a growth factor polypeptide corresponding to wild type human IGF-I (mature form).
  • SEQ ID NO:4 is the amino acid sequence of a growth factor polypeptide corresponding to human IGF-1 with D12A substitution.
  • SEQ ID NO:5 is the amino acid sequence of a growth factor polypeptide corresponding to human IGF-1 with E9A substitution.
  • SEQ ID NO:6 is the amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain N-K1 domain.
  • SEQ ID NO:7 is the amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain K1 domain.
  • SEQ ID NO:8 is the amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain N-K2 fusion.
  • SEQ ID NO:9 is the amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain K2 domain.
  • SEQ ID NO:10 is the amino acid sequence of a human serum albumin (HSA) linker with C34S and N503Q substitutions.
  • HSA human serum albumin
  • SEQ ID NO:11 is the nucleic acid sequence of an HSA linker with C34S and N503Q substitutions.
  • SEQ ID NO:12 is the amino acid sequence of HSA.
  • SEQ ID NO:13 is the nucleic acid sequence of HSA.
  • SEQ ID NO:14 is the amino acid sequence of an HSA linker with C34S and N503Q substitutions and a polypeptide connector.
  • SEQ ID NO:15 is the amino acid sequence of an HSA linker with C34S and N503Q substitutions and a polypeptide connector.
  • SEQ ID NO:16 is the amino acid sequence of an HSA linker with C34S and N503Q substitutions and a polypeptide connector.
  • SEQ ID NO:17 is the amino acid sequence of an HSA linker with C34S and N503Q substitutions and a polypeptide connector.
  • SEQ ID NO:18 is the amino acid sequence of an HSA linker with C34S and N503Q substitutions and a polypeptide connector.
  • SEQ ID NO:19 is the amino acid sequence of an HSA linker with a polypeptide connector.
  • SEQ ID NO:20 is the amino acid sequence of an HSA linker with a polypeptide connector.
  • SEQ ID NO:21 is the amino acid sequence of an HSA linker with a polypeptide connector.
  • SEQ ID NO:22 is the amino acid sequence of an HSA linker with a polypeptide connector.
  • SEQ ID NO:23 is the amino acid sequence of an HSA linker with a polypeptide connector.
  • SEQ ID NO:24 is the amino acid sequence of an HSA linker with C34S substitution, domain I.
  • SEQ ID NO:25 is the amino acid sequence of an HSA linker, domain II.
  • SEQ ID NO:26 is the amino acid sequence of an HSA linker with N503Q substitution, domain III.
  • SEQ ID NO:27 is the amino acid sequence of an HSA linker, domain I.
  • SEQ ID NO:28 is the amino acid sequence of an HSA linker, domain II.
  • SEQ ID NO:29 is the amino acid sequence of human alpha-fetoprotein.
  • SEQ ID NO:30 is the amino acid sequence of the anti-phosphatidylserine scFv PS4A7.
  • SEQ ID NO:31 is the amino acid sequence of human annexin V.
  • SEQ ID NO:32 is an amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain N-K1 domain.
  • SEQ ID NO:33 is an amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain K1 domain.
  • SEQ ID NO:34 is an amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain N-K2 domain.
  • SEQ ID NO:35 is an amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain K2 domain.
  • SEQ ID NO:36 is an amino acid sequence of a growth factor polypeptide corresponding to human VEGF alpha monomer.
  • SEQ ID NO:37 is an amino acid sequence of a growth factor polypeptide corresponding to human VEGF alpha dimer.
  • SEQ ID NO:38 is an amino acid sequence of a growth factor polypeptide corresponding to human FGF2.
  • SEQ ID NO:39 is an amino acid sequence of a growth factor polypeptide corresponding to human NRG1 alpha, EGF-like domain.
  • SEQ ID NO:40 is an amino acid sequence of a growth factor polypeptide corresponding to human NRG1 alpha, full sequence.
  • SEQ ID NO:41 is an amino acid sequence of a bi-specific fusion protein leader polypeptide.
  • SEQ ID NO:42 is an amino acid sequence of a bi-specific fusion protein leader polypeptide.
  • SEQ ID NO:43 is an amino acid sequence of a C-terminal hexahistidine-comprising polypeptide.
  • SEQ ID NO:44 is an amino acid sequence of a C-terminal hexahistidine-comprising polypeptide.
  • SEQ ID NO:45 is an amino acid sequence of a HSA linker.
  • SEQ ID NO:46 is an amino acid sequence of a HSA linker with N-terminal and C-terminal short connector polypeptides.
  • SEQ ID NO:47 is an amino acid sequence of a HSA linker with N-terminal and C-terminal short connector polypeptides.
  • SEQ ID NO:48 is an amino acid sequence of a HSA linker with N-terminal and C-terminal short connector polypeptides.
  • SEQ ID NO:49 is an amino acid sequence of a HSA linker with N-terminal and C-terminal short connector polypeptides.
  • the present invention is directed to bi-specific fusion proteins that comprise: (1) a targeting polypeptide domain that binds to an ischemia-associated molecule; and (2) an activator domain, such as a growth factor polypeptide or a cytokine polypeptide.
  • the bi-specific fusion protein further comprises: (3) a polypeptide linker having two termini, an N-terminus and a C-terminus, that is joined at one terminus via a peptide bond to the targeting polypeptide domain and is joined at the other terminus via a peptide bond to the activator domain.
  • bi-specific fusion proteins find use, for example, in recruiting cells that express one or more growth factor and/or cytokine (e.g., chemokine) receptors (e.g., stem cells, progenitor cells or immune system cells) to tissue following an ischemic event (e.g., to damaged cells).
  • cytokine e.g., chemokine
  • ischemic event e.g., to damaged cells.
  • the administration of such bi-specific fusion proteins may be used to facilitate repair or regeneration of damaged tissue.
  • polypeptide is used herein to refer to a molecule that consists of multiple amino acid residues linked by peptide bonds. This term carries no implication as to the number of amino acid residues so linked.
  • bi-specific refers to the ability of the fusion protein to interact with two different ligands: an ischemia-associated molecule (bound by the targeting polypeptide domain) and a receptor for the activator domain.
  • an ischemia-associated molecule bound by the targeting polypeptide domain
  • a receptor for the activator domain The binding properties of the targeting polypeptide domain and the activator domain are discussed in more detail below.
  • an “ischemia-associated molecule” is any molecule that is detected at a level that is significantly higher (e.g., at least 2-fold higher) following ischemia or hypoxia. Any suitable binding assay may be used to identify ischemia-associated molecules, including those provided herein.
  • the increased level of molecule that is detected may be the result of upregulation or decreased turnover, or may be due to increased accessibility (e.g., resulting from cell damage).
  • the ischemia-associated molecule is detected in a cell of post-ischemic tissue at a significantly higher level (e.g., at least 2-fold higher) than in a cell of the same tissue that has not undergone an ischemic event (i.e., the molecule is specific to or enriched in the post-ischemic tissue).
  • the ischemia-associated molecule is associated with cell damage (i.e., the molecule is detected at a significantly higher level in cells that are damaged than in undamaged cells of the same type).
  • Certain ischemia-associated molecules are enriched (2 fold or higher) in the heart after an ischemic event (or in a model system that is used to mimic ischemia in the heart).
  • Such molecules include, for example, molecules that are exposed on myocytes or other cardiac cells that undergo necrosis (such as DNA) or apoptosis (e.g., phosphatidylserine) or molecules that are enriched in scarred heart tissue, such as collagen (collagen I, III), myosin (including the cell type-specific subtypes thereof), or other extracellular matrix proteins that are enriched in post ischemic hearts.
  • Such molecules can be identified on the basis of enrichment following ischemia-reperfusion in vivo or in simulated ischemia-reperfusion in vitro, or following exposure to conditions such as hypoxia, decreased ATP, increased reactive oxygen species (ROS) or nitric oxide synthase (NOS) production, or serum starvation of cells cultured in vitro.
  • hypoxia decreased ATP
  • ROS reactive oxygen species
  • NOS nitric oxide synthase
  • binding to the ischemia-associated molecule is mediated by the targeting polypeptide domain.
  • This domain may be any polypeptide sequence that serves this function; in preferred embodiments, the targeting polypeptide domain comprises one or more antibody variable regions.
  • an “antibody” is a protein consisting of one or more polypeptides substantially encoded by immunoglobulin genes.
  • a typical antibody is a tetramer that is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kD) and one “heavy” chain (about 50-70 kD).
  • V L ” and V H refer to these light and heavy chains respectively.
  • An “antibody variable region” is an N-terminal region of an antibody variable chain (V L or V H ) comprising amino acid residues that are primarily responsible for antigen recognition. Those of ordinary skill in the art are readily able to identify an antibody variable region and to determine the minimum size needed to confer antigen recognition.
  • an antibody variable region comprises at least 70 amino acid residues, and more commonly at least 100 amino acid residues.
  • a polypeptide that comprises an antibody variable region may (but need not) further comprise other light and/or heavy chain sequences, and may (but need not) further comprise sequences that are not antibody-derived. It will be apparent that the sequence of an antibody variable region may be naturally-occurring, or may be modified using standard techniques, provided that the function (antigen recognition) is retained.
  • polypeptides that comprise an antibody variable region are single chain antibodies (antibodies that exist as a single polypeptide chain), more preferably single chain Fv antibodies (scFv) in which a variable heavy chain region and a variable light chain region are joined together (directly or through a peptide linker) to form a continuous polypeptide.
  • the scFv antibody may be chemically synthesized or may be expressed from a nucleic acid including V H - and V L -encoding sequences either joined directly or joined by a peptide-encoding linker.
  • Binding indicates that an antibody exhibits substantial affinity for a specific antigen (e.g., an ischemia-associated molecule) and is said to occur when the fusion protein (or the targeting polypeptide domain thereof) has a substantial affinity for the target antigen and is selective in that it does not exhibit significant cross-reactivity with other antigens.
  • Preferred substantial binding includes binding with a dissociation constant (K d ) of 10 ⁇ 6 , 10 ⁇ 7 , 10 ⁇ 8 , 10 ⁇ 9 , 10 ⁇ 10 , 10 ⁇ 11 , 10 ⁇ 12 M or better.
  • K d of an antibody-antigen interaction indicates the concentration of antibody (expressed as molarity) at which 50% of antibody and antigen molecules are bound together at thermodynamic equilibrium.
  • K d k off /k on .
  • affinities are stronger affinities, and are identified by dissociation constants of lower numeric value than their comparators, with a K d of 10 ⁇ 10 M being of lower numeric value and therefore representing a better affinity than a K d of 10 ⁇ 9 M. Affinities better (i.e., with a lower K d value and therefore stronger) than 10 ⁇ 7 M, preferably better than 10 ⁇ 8 M, are generally preferred.
  • preferred binding affinity can be indicated as a range of dissociation constants, for example preferred binding affinities for antibodies disclosed herein are represented by K d values ranging from 10 ⁇ 6 to 10 ⁇ 12 M (i.e., micromolar to picomolar), preferably 10 ⁇ 7 to 10 ⁇ 12 M, more preferably 10 ⁇ 8 to 10 ⁇ 12 M or better.
  • K d values ranging from 10 ⁇ 6 to 10 ⁇ 12 M (i.e., micromolar to picomolar), preferably 10 ⁇ 7 to 10 ⁇ 12 M, more preferably 10 ⁇ 8 to 10 ⁇ 12 M or better.
  • An antibody that “does not exhibit significant cross-reactivity” is one that will not appreciably bind to an off-target antigen.
  • an antibody that specifically and selectively binds to Annexin V will exhibit at least a two, and preferably three, or four or more orders of magnitude better binding affinity (i.e., binding exhibiting a two, three, or four or more orders of magnitude lower K d value) for Annexin V than for Annexin molecules other than Annexin V or for non-Annexin proteins or peptides.
  • Binding affinity and selectivity can be determined using any art-recognized methods for determining such characteristics, including, for example, using Scatchard analysis and/or competitive (competition) binding assays.
  • Binding may be assessed, and K d values determined, using any of a variety of techniques that are well known in the art. For example, binding to an ischemia-associated DNA molecule is commonly assessed by coating an appropriate solid support (e.g., beads, ELISA plate or BIACORE chip) with target DNA fragments. For a targeting polypeptide domain that binds to any sequence of DNA, DNA fragments (single or double-stranded) of 10 base pairs or larger are immobilized on the solid substrate. For a targeting polypeptide domain that binds to a specific sequence or DNA complex (e.g., DNA-histone complex) the appropriate corresponding target is immobilized.
  • an appropriate solid support e.g., beads, ELISA plate or BIACORE chip
  • non-specific binding sites for protein Prior to adding the ischemia-associated molecule, non-specific binding sites for protein are blocked with BSA, milk, or any other appropriate blocker. Uncoated wells or wells coated with a non-target molecule serve as specificity controls. Increasing concentrations of the bi-specific fusion protein (or targeting polypeptide domain) are incubated with target-coated substrate or control substrate. A fusion protein or domain that does not bind to the target is also tested as a specificity control. Target specific, dose-dependent binding of the bi-specific fusion protein (or targeting polypeptide domain) is then assessed by measuring the amount of bi-specific fusion protein (or targeting polypeptide domain) binding to target versus controls as a function of increasing dose using standard protocols corresponding to the solid support and binding technology being used.
  • binding affinity and kinetic on and off rates for binding to the target molecule are measured using standard techniques and compared to other negative control molecules (e.g., fusion protein with irrelevant targeting polypeptide or fusion protein lacking a targeting polypeptide) and positive control molecules (e.g., parental antibody that targets the ischemia-associated molecule, or other antibodies or antibody fragments that are known to bind to the ischemia-associated molecule).
  • negative control molecules e.g., fusion protein with irrelevant targeting polypeptide or fusion protein lacking a targeting polypeptide
  • positive control molecules e.g., parental antibody that targets the ischemia-associated molecule, or other antibodies or antibody fragments that are known to bind to the ischemia-associated molecule.
  • the K d is determined using a biosensor (e.g., by surface Plasmon resonance (BIAcore) or resonant mirror analysis (IAsys)). Such determinations may be performed as described by Hefta et al., Measuring Affinity Using Biosensors, in “Antibody Engineering: A Practical Approach,” McCafferty et al. (eds), pp. 99-116 (Oxford University Press, 1996), and references cited therein. Briefly, kinetic on and off rates (k on , and k off ) are determined using a sensor chip to which the ischemia-associated molecule has been coupled.
  • a biosensor e.g., by surface Plasmon resonance (BIAcore) or resonant mirror analysis (IAsys)
  • Such determinations may be performed as described by Hefta et al., Measuring Affinity Using Biosensors, in “Antibody Engineering: A Practical Approach,” McCafferty et al
  • a bi-specific fusion protein binds to the ischemia-associated molecule if it binds with a K d of less than 10 ⁇ 8 M, preferably less than 10 ⁇ 7 M, 10 ⁇ 8 M, 10 ⁇ 9 M or 10 ⁇ 10 M.
  • the binding of the bi-specific fusion protein to the ischemia-associated molecule in this assay is significantly higher (e.g., at least 2-, 10- or 100-fold higher) than binding of the bi-specific fusion protein to negative controls.
  • binding to the immobilized target can also be competed using excess soluble target.
  • ischemia-associated molecules are specific to (or enriched in) damaged cells. Binding to damaged cells is conveniently demonstrated in vitro using cultured cells that are exposed to conditions that induce necrosis or apoptosis. For example, necrosis can be induced in cultured cardiomyocytes by simulated ischemia/reperfusion, and monitored using a LDH release assay, or trypan blue assay followed by subtraction of the number of cells undergoing apoptosis, essentially as described in Shan et al., Am. J. Physiol. Cell. Physiol. 294:833-841 (2008).
  • This assay quantitates the total dead cells and the difference between the total and the number of apoptotic cells is attributed to necrosis, as discussed in more detail below.
  • Conditions that induce apoptosis include exposure to H 2 O 2 , and apoptosis can be monitored using any of a variety of techniques known in the art including, for example, annexin V (SEQ ID No. 31) binding cleavage of target peptide sequences by known caspases that are activated by apoptosis, or DNA laddering (measured by TUNEL assay, essentially as described in Kuramochi, J. Biol. Chem. 279(49): 51141-47 (2004)).
  • Binding to the cells undergoing necrosis or apoptosis may be assessed by adding fluorescently labeled bi-specific fusion protein (or targeting polypeptide domain) or appropriate control proteins to cells following the induction of apoptosis or necrosis. After incubation of the proteins with the cells for times ranging from a few minutes to one day, the cells are washed and then the cell-bound fluorescence is measured using immunofluorescence, flow cytometry, or similar techniques.
  • bi-specific fusion protein or targeting polypeptide domain
  • methods of detecting the bound bi-specific fusion protein (or targeting polypeptide domain) including radiolabeling or using enzymes conjugated to the bi-specific fusion protein (or targeting polypeptide domain) or to antibodies that bind to the fusion protein (or targeting polypeptide domain), which is common practice in ELISA protocols.
  • the bi-specific fusion protein (or targeting polypeptide domain) binds to target cells if significantly higher (e.g., 2-fold higher) binding to cells following ischemia (e.g., cells undergoing necrosis or apoptosis) is detected, as compared to cells that have not experienced an ischemic event (e.g., cells not undergoing apoptosis or necrosis).
  • In vivo targeting may be demonstrated by inducing ischemia in an animal model and comparing the level of administered bi-specific fusion protein (or targeting polypeptide domain) in a target tissue before and after ischemia.
  • In vivo targeting to damaged cells may be demonstrated by inducing tissue damage in an animal model, administering the bi-specific fusion protein (or targeting polypeptide domain), and comparing the level of bi-specific fusion protein (or targeting polypeptide domain) in damaged versus undamaged cells.
  • the bi-specific fusion proteins are designed to target areas of tissue damage following ischemia-reperfusion injury.
  • demonstration of in vivo targeting may be accomplished by inducing tissue damage, preferably by a method that causes ischemia followed by re-establishment of blood supply.
  • tissue damage preferably by a method that causes ischemia followed by re-establishment of blood supply.
  • Numerous methods are available to do this in different tissues.
  • blood flow to the hindlimb of the mouse can be transiently blocked with a simple tourniquet.
  • temporary clamp on the artery leading into the kidney can be employed.
  • Ischemia-reperfusion injury can be induced in the heart through temporary blockage of the coronary artery as demonstrated in mice, rats, dogs, and pigs. Representative methods for inducing tissue damage in an animal model are summarized in Table 1.
  • the specificity of targeting can be established by comparing the bi-specific fusion protein (or targeting polypeptide domain) deposition in the clamped versus unclamped kidney as shown in Chen et al., FASEB J. 4(12): 3033-39 (1990), or in the treated versus untreated hindlimb as shown in Zbinden et al., Am. J. Physiol. Heart Circ. Physiol. 292: H1891-H1897 (2007), using radiolabeled bi-specific fusion protein (or targeting polypeptide domain).
  • bi-specific fusion protein (or targeting polypeptide domain) can be detected in homogenized tissue using ELISA, or can be imaged in real time using bi-specific fusion protein (or targeting polypeptide domain) labeled with the appropriate metal for imaging (e.g., Tc99, Y or Gd).
  • the appropriate metal for imaging e.g., Tc99, Y or Gd.
  • Specific deposition in the damaged area of the heart can be measured as described in Dumont et al., Circulation 102(13):1564-8 (2000). Representative methods for demonstrating targeting of proteins to damaged tissue are shown in Table 2.
  • certain targeting polypeptide domains comprise a scFv antibody that binds to the ischemia-associated molecule.
  • Representative such scFv antibodies comprise or have the sequences provided herein as SEQ ID NOs: 1, 2, and 30.
  • Antibodies may also, or alternatively, be used as a targeting polypeptide domain.
  • Antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain complementarity determining regions (CDRs). For this reason, the amino acid sequences within CDRs are more diverse between individual antibodies than sequences outside of CDRs. Because CDR sequences are responsible for most antibody-antigen interactions, it is possible to generate modified antibodies that mimic the properties of an original antibody by combining CDR sequences from one antibody with framework sequences from a different antibody.
  • Such framework sequences can be obtained from public DNA databases that include germline antibody gene sequences.
  • one or more CDRs of a targeting polypeptide domain sequence provided herein can be used to create functionally related antibodies that retain the binding characteristics of the original targeting polypeptide domain.
  • one or more CDR regions selected from SEQ ID NOs: 1, 2, and 30, is combined recombinantly with known human framework regions and CDRs to create additional, recombinantly-engineered, targeting polypeptide domains.
  • the heavy and light chain variable framework regions can be derived from the same or different antibody sequences. CDR regions are readily identified using alignments with known sequences in databases such as Vbase and IMGT.
  • the resulting targeting polypeptide domains share one or more CDRs with the targeting polypeptide domains of SEQ ID NOs: 1, 2, and 30; in certain embodiments, the targeting polypeptide domain comprises at least one CDR of a sequence as recited in SEQ ID NO: 1, 2, or 30.
  • antibodies are generated that include the heavy and/or light chain CDR3s of the particular antibodies described herein.
  • the antibodies can further include the heavy and/or light chain CDR1 and/or CDR2s of the antibodies disclosed herein.
  • the CDR 1, 2, and/or 3 regions of the engineered antibodies described above can comprise the exact amino acid sequence(s) as those disclosed herein.
  • the ordinarily skilled artisan will appreciate that some deviation from the exact CDR sequences may be possible, particularly for CDR1 and CDR2 sequences, which can tolerate more variation than CDR3 sequences without altering epitope specificity (such deviations are, e.g., conservative amino acid substitutions).
  • the engineered antibody may be composed of one or more CDR1s and CDR2s that are, for example, 90%, 95%, 98%, 99% or 99.5% identical to the corresponding CDRs of an antibody named herein.
  • one or more residues of a CDR may be altered to modify binding to achieve a more favored on-rate of binding.
  • Affinity maturation techniques can be used to alter the CDR region(s) followed by screening of the resultant binding molecules for the desired change in binding. Accordingly, as CDR(s) are altered, changes in binding affinity as well as immunogenicity can be monitored and scored such that an antibody optimized for the best combined binding and low immunogenicity are achieved.
  • Modifications can also be made within one or more of the framework or joining regions of the heavy and/or the light chain variable regions of an antibody, so long as antigen binding affinity subsequent to these modifications is not substantially diminished.
  • the activator domain is any polypeptide that detectably modulates the activity of a cellular network; certain activator domains are growth factor polypeptides or cytokine polypeptides (e.g., a chemokine polypeptide). It will be apparent that such modulation may be an increase or a decrease in the activity of the cellular network.
  • a growth factor polypeptide detectably modulates activation of a growth factor receptor (such as HGF or IGF receptor).
  • Certain such polypeptides are wild-type hepatocyte growth factor (HGF) or HGF alpha chain (e.g., GENBANK accession number P14210), or derivatives thereof that retain at least 10% of wild-type biological activity, as determined by measuring activation of the corresponding growth factor receptor in appropriate target cells.
  • Activation may be assessed, for example, by measuring phosphorylation of receptor kinase or downstream proteins, such as AKT, essentially as described by Nishi et al., Proc. Natl. Acad. Sci. USA 95:7018-7023 (1998). MTT and CTG assays known in the art may also be used.
  • Representative growth factor polypeptides have a sequence as recited in SEQ ID NO:3-9 or 32-40, herein.
  • activator domains that share one or more CDRs with the activator domains of SEQ ID NOs: 3-9 or 32-40 are also contemplated; CDRs may be identified and such activator domains may be constructed using well known techniques.
  • the activator domain comprises at least one CDR of a sequence as recited in SEQ ID NO:3-9 or 32-40.
  • a cytokine polypeptide modulates activation of the corresponding cytokine receptor, as determined in the same fashion.
  • the activator domain is a growth factor polypeptide, which binds a growth factor receptor on a cell surface.
  • growth factor receptors are receptors for epidermal growth factor (EGF), Neregulin/Heregulin (NRG), fibroblast growth factor (FGF), insulin-like growth factor (e.g., IGF-I), platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF) and isoforms thereof (e.g., VEGF-A or VEGF-C), teratocarcinoma-derived growth factor 1 (TDGF1), transforming growth factor alpha (TGF- ⁇ ), transforming growth factor beta (TGF- ⁇ ) and isoforms thereof (e.g., TGF- ⁇ 1 or TGF-( ⁇ 2), thrombopoietin (THPO) or periostin.
  • EGF epidermal growth factor
  • NSG Neregulin/Heregulin
  • FGF fibroblast growth factor
  • IGF-I
  • SCFR mast/stem cell growth factor receptor
  • HGF hepatocyte growth factor receptor
  • ErbB-3 ErbB-4
  • high affinity nerve growth factor receptor BDNF/NT-3 growth factors receptor
  • NT-3 growth factor receptor NT-3 growth factor receptor
  • VEGFR-I vascular endothelial growth factor receptor 1
  • cytokine receptors include, for example, FL cytokine receptor, receptor for cytokine receptor common gamma chain, interleukin-10 receptor alpha chain, interleukin-10 receptor beta chain, interleukin-12 receptor beta-1 chain, interleukin-12 receptor beta-2 chain, interleukin-13 receptor alpha-1 chain, interleukin-13 receptor alpha-2 chain, interleukin-17 receptor; interleukin-17B receptor, interleukin 21 receptor precursor, interleukin-1 receptor type I, interleukin-1 receptor type II, interleukin-2 receptor alpha chain, interleukin-2 receptor beta chain, interleukin-3 receptor alpha chain, interleukin-4 receptor alpha chain, interleukin-5 receptor alpha chain, interleukin-6 receptor alpha chain, interleukin-6 receptor beta chain, interleukin-7 receptor alpha chain, high affinity interleukin-8 receptor A, high affinity interleukin-8 receptor B, interleukin-9 receptor, interleukin-18 receptor 1, interleukin-1 receptor-like
  • Still other activator domains are receptors for solute carrier organic anion transporter family, member 1A2 (SLCO1A2), sphingosine kinase 1 (SPHK1), secreted phosphoprotein 1 (SPP1), also called osteopontin (OPN), tumor protein 53 (TP53), troponin T type 1 (TNNT1), TSPY-like protein 2 (TSPYL2), visfatin, WAP four-disulfide core domain 1 (WFDC1), thymosin beta 4, wingless-type MMTV integration site family, member 11 (WNT11).
  • SCO1A2 solute carrier organic anion transporter family
  • SPHK1 sphingosine kinase 1
  • SPP1 secreted phosphoprotein 1
  • OPN osteopontin
  • TP53 tumor protein 53
  • TNNT1 troponin T type 1
  • TSPY-like protein 2 TSPY-like protein 2
  • WFDC1 WAP four-disulfide
  • activator domains include, for example, resistin, stromal cell-derived factor-1 (SDF-1), signal-induced proliferation-associated gene 1 (SIPA1), and any of the other ligands listed above, as well as portions and derivatives of the foregoing that substantially retain the ability to bind to cognate receptors.
  • SDF-1 stromal cell-derived factor-1
  • SIPA1 signal-induced proliferation-associated gene 1
  • binding of a bi-specific fusion protein (or activator domain thereof) to the appropriate receptor may be assessed using techniques known in the art.
  • binding is demonstrated by coating an appropriate solid support with the recombinant ectodomain of the appropriate receptor.
  • An ectodomain from a receptor not recognized by the activator domain of the bi-specific fusion protein is used as a specificity control.
  • a support substrate that does not have any immobilized receptor is also used as a control. Similar to the methods described above for binding to the ischemia-associated molecule, specific, dose-dependent binding to receptor is demonstrated using standard protocols corresponding to the solid support and binding technology being used.
  • bi-specific fusion protein is immobilized to a support and the binding of the soluble ectodomain of the corresponding receptor(s) is used to demonstrate dose-dependent, specific binding.
  • the binding affinity and kinetic on and off rates for binding of the bi-specific fusion protein to the receptor(s) are also measured using standard techniques and compared to other negative control molecules (fusion protein with irrelevant control activator domain, fusion protein lacking an activator domain) and positive control molecules (recombinant wild-type receptor ligand, such as a growth factor or cytokine).
  • the equilibrium and kinetic binding parameters of the bi-specific fusion protein are also compared to the same parameters measured for the un-fused wild-type ligand to determine whether fusion of the ligand to other molecules affects the normal binding of the ligand to its corresponding receptor. Such information may be used to determine the effective dose of the bi-specific fusion protein.
  • a bi-specific fusion protein binds to immobilized growth factor receptor or cytokine receptor with a significantly higher affinity (e.g., at least 100-fold) than that observed for negative controls.
  • binding to the immobilized receptor can be competed using excess soluble polypeptide, soluble receptor, or antibodies that bind to polypeptide or receptor and block their interaction.
  • the bi-specific fusion protein binds to the growth factor or cytokine receptor with an affinity within 1000-fold of the native ligand binding to its receptor.
  • a bi-specific fusion protein (and its activator domain) further has the capacity to mediate cognate receptor activation.
  • Such activity may be assessed, for example, using a cellular model of ischemia reperfusion, which uses cultured cardiomyocytes such as neonatal rat ventricular myocytes (NRVM) or cell lines.
  • Simulated ischemia (SI) is generally initiated by metabolic inhibitors (deoxyglucose and dithionite) and metabolites (high potassium, lactate, low pH) or by hypoxia in an anaerobic chamber. Reperfusion is simulated by resuspension in an oxygenated buffer.
  • Native growth factors and cytokines can be used as activator domains. It will be apparent, however, that portions of such native sequences and polypeptides having altered sequences may also be used, provided that such polypeptides retain the ability to activate the cognate receptor (e.g., using one of the assays discussed below, such polypeptides detectably activate the receptor, and preferably activate the receptor to a degree that is at least 1% (preferably at least 10%) of that observed for the native ligand.
  • Certain activator domains that bind to growth factor receptors are provided herein in SEQ ID NOs:3-9 and 32-40. Activity of fusion proteins comprising such sequences is well known in the art (e.g., Hashino et al., J. Biochem. 119(4):604-609 (1996); Nishi et al., Proc. Natl. Acad. Sci. USA 95:7018-23 (1998)).
  • An activator domain for a particular application may be selected based on the desired therapeutic outcome.
  • an activator domain that comprises FGF2, VEGF alpha or a portion or derivative thereof that substantially retains the ability to bind to cognate receptor may generally be used to increase angiogenesis.
  • activator domains that comprise IGF, HGF or NRG1 may be used.
  • an ELISA may be conveniently used for this purpose.
  • the substrate of the targeting polypeptide e.g., DNA
  • the bi-specific fusion protein is then added, followed by addition of recombinant substrate for the activator domain (e.g., if the activator is a growth factor, then the substrate is recombinant cognate receptor or receptor fragment (ectodomain)).
  • This substrate is either fluorescently labeled for detection or detected using a labeled antibody to a region of the receptor that does not significantly affect ligand binding.
  • the in vivo activity of the bi-specific fusion protein is generally assessed by detecting signaling changes in molecules that are regulated by the activator domain of the bi-specific fusion protein. This typically involves changes in cell surface receptor phosphorylation status or downstream mediators such as phospho-AKT or phospho-ERK as detected by flow cytometry, immunofluorescence, ELISA, phospho-labeling, or Western analysis of treated tissues. Other functional assessments include tests for the number of viable cells by staining and morphological identification, level of apoptosis by Annexin V binding (via immunofluorescence) or flow cytometry, detection of caspase activity, TUNEL-assay (reduced number of TUNEL-positive cells) or DNA laddering. In each case, a bi-specific fusion protein functions in vivo if it induces a significant (e.g., at least 2-fold) change in the level, functional activity or phosphorylation of the regulated molecule detected by the assay.
  • a bi-specific fusion protein
  • repair of damaged tissue in a patient can be assessed using any clinically relevant standard.
  • repair of infracted tissue can be measured by quantitation of cell number, such as the number of myocytes, fibroblast, or amount of scarring, or with functional assays for output or structural aspects of heart function including, LVEDP, LVDP, +dp/dT, LV Weight, Chamber Volume, and Diastolic Wall Stress.
  • LVEDP low-density
  • LVDP LVDP
  • +dp/dT LV Weight
  • Chamber Volume Chamber Volume
  • Diastolic Wall Stress e.g., a bi-specific fusion protein is said to repair damaged tissue if it results in a significant (e.g., at least 2-fold) change in any such clinical assessment.
  • the targeting polypeptide domain and activator domain may be directly joined via a peptide bond. Alternatively, they may be joined via a polypeptide linker. It will be apparent that any such linker will have two termini, an N-terminus and a C-terminus.
  • the linker is joined at one terminus via a peptide bond to the targeting polypeptide domain and is joined at the other terminus via a peptide bond to the activator domain.
  • the linker is joined at the N-terminus to the C-terminus of the targeting polypeptide domain and at the C-terminus to the N-terminus of the activator domain.
  • the linker is joined at the C-terminus to the targeting polypeptide domain and at the N-terminus to the activator domain.
  • the linker is non-immunogenic in humans. More preferably, the linker is a human serum protein or a derivative thereof that retains at least 50% sequence identity over a region that consists of at least 100 consecutive amino acids. In further embodiments, the linker comprises at least 100 consecutive amino acids that are at least 70%, 80%, 85%, 90% or 95% identical to a human serum albumin amino acid sequence or a human alpha-fetoprotein amino acid sequence. Representative such linkers include those recited in any one of SEQ ID NOs:10, 12, 14-29 and 45, which may be incorporated into a bi-specific fusion protein alone or using a short (e.g., from 2 to 20 amino acid residues) connector polypeptide at one or both ends.
  • Suitable short connector polypeptides for use at the N-terminal end of the linker include, for example, dipeptides such as -Gly-Ala-(GA) and -Ala-Ser-(AS).
  • Suitable short connector polypeptides for use at the C-terminal end of the linker include, for example, dipeptides such as -Leu-Gln-(LQ) and -Thr-Gly-(TG).
  • SEQ ID NOs:46-49 recite the linker of SEQ ID NO:45 with representative connector dipeptides at both the N- and C-termini; it will be apparent, however, that such short connector polypeptides, if present, may be located at either one or both termini.
  • Certain preferred linkers provide a prolonged half-life of the bi-specific fusion protein, as compared to fusion protein without linker.
  • the effect of a linker on half-life can be evaluated using an assay that determines stability under physiological conditions.
  • bi-specific fusion protein can be incubated at 37° C. in serum (e.g., human) for 120 hours, with samples removed at the start of incubation and every 24 hours thereafter. Binding assays as described above are then performed to detect the level of functional bi-specific fusion protein at each time point. This level is then compared to the level of bi-specific fusion protein constructed without linker (or using a different linker) to provide a half-life comparison.
  • bi-specific fusion proteins may optionally be included in the bi-specific fusion proteins provided herein. Such elements may be present for a variety of purposes, including to facilitate expression, preparation or purification of the bi-specific fusion protein, or to perform targeting functions.
  • an N-terminal leader polypeptide may be present. Representative leader polypeptides comprise or have a sequence recited in SEQ ID NO:41 or 42.
  • a bi-specific fusion protein may also, or alternatively, comprise a polyhistidine (e.g., hexahistidine) tag to facilitate purification. Such a tag comprises at least six histidine consecutive amino acid residues, and may be located at the C- or N-terminus.
  • a hexahistidine tag is included at the C-terminus of the bi-specific fusion protein. Additional amino acid residues may also be present at the junction of the polyhistidine to the remainder of the bi-specific fusion protein.
  • Certain bi-specific fusion proteins provided herein comprise a C-terminal polyhistidine-comprising polypeptide as recited in SEQ ID NO:43 or 44.
  • Certain bi-specific fusion proteins have a general structure that satisfies one of the following (shown from N-terminal to C-terminal, left to right):
  • bi-specific fusion proteins comprise (from N-terminal to C-terminal):
  • leader polypeptide e.g., comprising or having a sequence recited in SEQ ID NO:41 or 42
  • a targeting polypeptide domain e.g., comprising or having a sequence recited in SEQ ID NO: 1, 2, 30 or 31;
  • a short connector polypeptide e.g., comprising or having the sequence Gly-Ala- or -Ala-Ser-;
  • HSA polypeptide e.g., comprising or having a sequence recited in any one of SEQ ID NOs:10, 12, 14-29 and 45;
  • a short connector polypeptide e.g., comprising or having the sequence -Leu-Gln- or -Thr-Gly-;
  • an activator domain e.g. comprising or having a sequence recited in any one of SEQ ID NOs:3-9 and 32-40;
  • a polyhistidine-comprising polypeptide e.g., a hexahistidine-comprising polypeptide, such as a polypeptide comprising or having a sequence recited in SEQ ID NO:43 or 44.
  • certain such bi-specific fusion proteins comprise (N-terminal to C-terminal): a leader sequence as recited in SEQ ID NO:41 or 42; a targeting polypeptide domain as recited in SEQ ID NO:1, 2, 30 or 31; an HSA polypeptide having the sequence recited in SEQ ID NO:45; a -Gly-Ala or Ala-Ser- connector dipeptide; -Leu-Gln- or -Thr-Gly-; an activator domain having a sequence recited in any one of SEQ ID NOs: 32-40; and a hexahistidine-comprising polypeptide having a sequence recited in SEQ ID NO:43 or 44.
  • bi-specific fusion proteins comprise (from N-terminal to C-terminal):
  • leader polypeptide e.g., comprising or having a sequence recited in SEQ ID NO:41 or 42
  • an activator domain e.g., comprising or having a sequence recited in any one of SEQ ID NOs:3-9 and 32-40;
  • a short connector polypeptide e.g., comprising or having the sequence -Gly-Ala- or -Ala-Ser-);
  • HSA polypeptide e.g., comprising or having a sequence recited in any one of SEQ ID NOs:10, 12, 14-29 and 45;
  • a short connector polypeptide e.g., comprising or having the sequence -Leu-Gln- or -Thr-Gly-;
  • a targeting polypeptide domain e.g., comprising or having a sequence recited in SEQ ID NO: 1, 2, 30 or 31;
  • poly-histidine-comprising polypeptide e.g., comprising or having as sequence recited in SEQ ID NO:43 or 44.
  • bi-specific fusion proteins comprise (from N-terminal to C-terminal):
  • leader polypeptide e.g., comprising or having a sequence recited in SEQ ID NO:41 or 42
  • an activator domain e.g., comprising or having a sequence recited in any one of SEQ ID NOs:3-9 and 32-40;
  • a targeting polypeptide domain e.g., comprising or having a sequence recited in SEQ ID NO: 1, 2, 30 or 31;
  • poly-histidine-comprising polypeptide e.g., comprising or having as sequence recited in SEQ ID NO:43 or 44.
  • Bi-specific fusion proteins may be synthesized using standard techniques, including liquid- and solid-phase peptide synthesis and recombinant DNA techniques.
  • solid phase synthesis the C-terminal amino acid of the sequence is attached to an insoluble support, and the remaining amino acids are added in sequence.
  • shorter regions may be synthesized in this fashion and then condensed to form the longer polypeptide.
  • DNA encoding the bi-specific fusion protein is prepared chemically or by isolating and ligating DNA encoding each portion of the fusion protein.
  • the DNA coding for each segment of the bi-specific fusion protein may be isolated from known genes or synthesized de novo. Methods for direct chemical synthesis of DNA are well known in the art, and such syntheses are routinely performed using an automated synthesizer. Chemical synthesis produces a single stranded polynucleotide, which is converted into double stranded DNA by hybridization with a complementary sequence or using DNA polymerase.
  • DNA sequences encoding the bi-specific fusion protein are prepared by cloning. Cloning techniques are well known in the art, and are amply described, for example, by standard references such as Sambrook et al., Molecular Cloning: A Laboratory Manual (3 rd ed.), Cold Spring Harbor Laboratory Press (2001). Portions of the DNA may be ligated together in frame to generate the full length coding sequence.
  • the DNA may be cloned into a vector for expression in a prokaryotic or eukaryotic host cell.
  • a vector for expression e.g., a prokaryotic or eukaryotic host cell.
  • the DNA encoding the bi-specific fusion protein is operably linked to the nucleotide sequences necessary for expression (e.g., a suitable promoter and, if necessary, a terminating signal).
  • a promoter is a nucleotide sequence (typically located 5′ to the coding sequence) that directs the transcription of adjacently linked coding sequences.
  • a terminating signal may be a stop codon to end translation and/or a transcription termination signal.
  • Additional regulatory element(s) may also be present within an expression vector.
  • a vector is preferably a plasmid or viral vector.
  • an expression vector further comprises a selectable marker, which confers resistance to a selection. This allows cells to stably integrate the vector into their chromosomes and grow to form foci, which in turn can be cloned and expanded into cell lines.
  • selectable markers are known in the art, including, for example, genes that provide resistance to ampicillin, methotrexate, mycophenolic acid, the aminoglycoside G-418, hygromycin and puromycin.
  • Host cells are transformed or transfected with the vector that comprises the DNA encoding the bi-specific fusion protein using standard methods. Expression in the host cell results in transcription of the DNA into the corresponding mRNA, followed by translation of the mRNA to generate the bi-specific fusion protein.
  • the bi-specific fusion protein can be purified according to standard procedures, including, for example, ammonium sulfate precipitation or affinity column chromatography. Substantially pure compositions of at least about 90 to 95% homogeneity are preferred, and 98 to 99% or more homogeneity is most preferred for pharmaceutical uses. Once purified, partially or to homogeneity as desired, if to be used therapeutically, the polypeptides should be substantially free of endotoxin.
  • the present invention also provides pharmaceutical compositions comprising at least one bi-specific fusion protein as described herein, together with at least one physiologically acceptable carrier.
  • Such compositions may be used for treating patients who are suffering from, or at risk for, tissue damage, in order to prevent tissue damage, or to repair or regenerate damaged tissue.
  • Such patients include, for example, patients who have experienced myocardial infarction, kidney damage, and/or ischemic stroke).
  • other active ingredients may also be included within the pharmaceutical composition, such as stem cells or other agents that facilitate repair of damaged tissue.
  • physiologically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the bi-specific fusion protein is administered.
  • Physiologically acceptable carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin (e.g., peanut oil, soybean oil, mineral oil, or sesame oil). Water is a preferred carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • suitable pharmaceutical excipients include, for example, starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water and ethanol.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • compositions may be formulated for any appropriate manner of administration, including, for example, parenteral, intranasal, topical, oral, or local administration, such as by a transdermal means, for prophylactic and/or therapeutic treatment.
  • These compositions can take any of a variety of well known forms that suit the mode of administration, such as solutions, suspensions, emulsions, tablets, pills, capsules, powders, aerosols and sustained-release formulations.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical modes of administration and carriers are described in “Remington: The Science and Practice of Pharmacy,” A. R. Gennaro, ed. Lippincott Williams & Wilkins, Philadelphia, Pa. (21 st ed., 2005
  • the pharmaceutical compositions provided herein are administered parenterally (e.g., by intravenous, intramuscular, or subcutaneous injection), or by oral ingestion or topical application.
  • parenteral administration the bi-specific fusion protein can either be suspended or dissolved in the carrier.
  • a sterile aqueous carrier is generally preferred, such as water, buffered water, saline or phosphate-buffered saline.
  • sterile, fixed oils may be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed, including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectible compositions.
  • auxiliary substances may also be included to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, dispersing agents, suspending agents, wetting agents, detergents, preservatives, local anesthetics and buffering agents.
  • the pharmaceutical composition is formulated for intravenous administration to a patient (e.g., a human).
  • a patient e.g., a human
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a sealed (e.g., hermetically sealed) container such as an ampoule or sachette indicating the quantity of active agent.
  • composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • compositions intended for oral use may be presented as, for example, tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. Such compositions may further comprise one or more components such as sweetening agents flavoring agents, coloring agents and preserving agents. Tablets contain the active ingredient in admixture with physiologically acceptable excipients that are suitable for the manufacture of tablets. Such excipients include, for example, inert diluents, granulating and disintegrating agents, binding agents and lubricating agents.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium.
  • Aqueous suspensions comprise the active materials in admixture with one or more excipients suitable for the manufacture of aqueous suspensions. Such excipients include suspending agents and dispersing or wetting agents. Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil (e.g., arachis oil, olive oil, sesame oil or coconut oil) or in a mineral oil such as liquid paraffin.
  • Pharmaceutical compositions may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil or a mineral oil or mixture thereof.
  • Suitable emulsifying agents include, for example, naturally-occurring gums, naturally-occurring phosphatides and anhydrides.
  • compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered. Sterile aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of an aqueous pharmaceutical composition typically will be between 3 and 11, more preferably between 5 and 9 or between 6 and 8, and most preferably between 7 and 8, such as 7 to 7.5.
  • Bi-specific fusion proteins provided herein are generally present within a pharmaceutical composition at a concentration such that administration of a single dose to a patient delivers a therapeutically effective amount.
  • a therapeutically effective amount is an amount that results in a discernible patient benefit, such as detectable repair or regeneration of damaged tissue or diminution of symptoms of tissue damage.
  • Therapeutically effective amounts can be approximated from the amounts sufficient to achieve detectable tissue repair or regeneration in one or more animal models exemplified in Table 3.
  • dosage unit forms contain between from about 10 g to about 100 g of bi-specific fusion protein.
  • compositions may be packaged for treating or preventing tissue damage (e.g., for treatment of myocardial infarction or kidney damage).
  • Packaged pharmaceutical preparations include a container holding a therapeutically effective amount of at least one pharmaceutical composition as described herein and instructions (e.g., labeling) indicating that the contained composition is to be used for treating tissue damage (such as myocardial infarction or kidney damage) in a patient.
  • Pharmaceutical compositions may be packaged in multiple single dose units, each containing a fixed amount of bi-specific fusion protein in a sealed package. Alternatively, the container may hold multiple doses of the pharmaceutical composition.
  • the pharmaceutical compositions can be administered to a patient (preferably a mammal such as a cow, pig, horse, chicken, cat, dog, or more preferably a human) to treat pathological tissue damage in the patient.
  • a patient preferably a mammal such as a cow, pig, horse, chicken, cat, dog, or more preferably a human
  • treatment encompasses both prophylactic and therapeutic administration.
  • a pharmaceutical composition as described herein is administered to a patient susceptible to or otherwise at risk for developing pathological tissue damage, in order to prevent, delay or reduce the severity of tissue damage.
  • treatment is performed in order to reduce the severity of the pathological tissue damage exist in the patient prior to treatment.
  • Representative pathological tissue damage includes heart tissue damage (e.g., damage associated with myocardial infarction), kidney tissue damage and ischemic stroke.
  • bi-specific fusion protein including, for example, encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the bi-specific fusion protein, receptor-mediated, or a retroviral or other nucleic acid vector.
  • the bi-specific fusion protein may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.), and may be administered together with other biologically active agents. Administration can be systemic or local.
  • intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • bsBAs of the invention may be desirable to administer the bsBAs of the invention locally to the area in need of treatment; this may be achieved by, for example, local infusion during surgery, topical application (e.g., in conjunction with a wound dressing after surgery), by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • a vesicle such as a liposome, can be used to deliver the bi-specific fusion protein.
  • the bi-specific fusion protein is delivered in a controlled release system; for example, such a controlled release system may be placed at or near the therapeutic target (e.g., an organ of the body that has experienced or is at risk for tissue damage).
  • a controlled release system may be placed at or near the therapeutic target (e.g., an organ of the body that has experienced or is at risk for tissue damage).
  • the therapeutic target e.g., an organ of the body that has experienced or is at risk for tissue damage.
  • the bi-specific fusion proteins provided herein are effective for treating pathological tissue damage at least in part due to their ability to recruit stem cells to the damaged tissue.
  • sufficient stem cells may reside within the patient (e.g., resident cardiac stem cells).
  • stem cells e.g., bone marrow-derived autologous stem cells.
  • Such stem cells may be administered before or after the bi-specific fusion protein, or may be administered simultaneously (either in the same pharmaceutical composition or in separate compositions).
  • the optimal dose depends on certain factors known in the art, but generally ranges from about 0.5 mg to about 400 mg of bi-specific fusion protein per dose (e.g., 10 mg, 50 mg, 100 mg, 200 mg, 300 mg, or 400 mg per dose).
  • a dose of bi-specific fusion protein (within a pharmaceutical composition as described above) can be administered therapeutically to a patient one or more times per hour, day, week, month, or year (e.g., 2, 4, 5, 6, 7, 8, 9, 10, 11, or 12 times per hour, day, week, month, or year). More commonly, a single dose per day or per week comprising an amount of bi-specific fusion protein ranging from about 0.1 g to about 100 g per kilogram of body weight is administered.
  • a pharmaceutical composition comprising a bi-specific fusion protein may be administered to a patient in a dosage that ranges from about 0.5 mg per week to about 400 mg per week, about 1.0 mg per week to about 300 mg per week, about 5 mg per week to about 200 mg per week, about 10 mg per week to about 100 mg per week, about 20 mg per week to about 80 mg per week, about 100 mg per week to about 300 mg per week, or about 100 mg per week to about 200 mg per week.
  • a pharmaceutical composition comprising a bi-specific fusion protein may be administered at a dose that ranges from about 0.5 mg every other day to about 100 mg every other day, about 5 mg every other day to about 75 mg every other day, about 10 mg every other day to about 50 mg every other day, or about 20 mg every other day to about 40 mg every other day.
  • a pharmaceutical composition comprising a bi-specific fusion protein may alternatively be administered at a dose that ranges from about 0.5 mg three times per week to about 100 mg three times per week, about 5 mg three times per week to about 75 mg three times per week, about 10 mg three times per week to about 50 mg three times per week, or about 20 mg three times per week to about 40 mg three times per week.
  • a pharmaceutical composition comprising a bi-specific fusion protein is administered to a mammal (e.g., a human) continuously for 1, 2, 3, or 4 hours; 1, 2, 3, or 4 times a day; every other day or every third, fourth, fifth, or sixth day; 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times a week; biweekly; 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 times a month; bimonthly; 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times every six months; 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 times a year; or biannually.
  • a pharmaceutical composition comprising a bi-specific fusion protein may, but need not, be administered at different frequencies during a therapeutic regime.
  • a bi-specific fusion protein in which targeting polypeptide domain binds to DNA and the activator domain is NRG1 is prepared.
  • the two domains are joined by a modified human serum albumin (HSA) linker.
  • HSA human serum albumin
  • the NRG1 is recombinantly fused to the amino terminus of the HSA linker incorporating a short connector polypeptide and the anti-DNA scFv is recombinantly fused to the carboxy terminus of the modified HSA linker incorporating an additional short connector polypeptide.
  • the modified HSA linker contains two amino acid substitutions.
  • a cysteine residue at position 34 of native HSA is mutated to serine in order to reduce potential protein heterogeneity due to oxidation at this site.
  • HSA linker confers an extended circulating half-life on the bi-specific fusion protein.
  • Example 1 The activity of both components of the representative bi-specific fusion protein prepared in Example 1 (in which the targeting polypeptide domain binds to DNA and the activator domain is NRG1) are tested using an ELISA designed to give activity only when both arms of the bi-specific fusion protein are bound to their substrates simultaneously.
  • the ELISA is performed essentially as described in Stokes et al., J. Clin. Pathol. 35(5): 566-573 (1982) and Gripenberg et al., Scand. J. Immunol. 1:151-157 (1978).
  • the amount of signal detected is significantly higher (at least 100-fold higher) in the wells with bi-specific fusion protein than in wells without DNA or negative controls that contain a dead arm (i.e., does not contain an activator domain or targeting polypeptide domain).
  • the signal is seen to vary with the amount of bi-specific fusion protein added to the wells.
  • the in vivo activity of the representative bi-specific fusion protein prepared in Example 1 is determined by detecting signaling changes in a molecule that is regulated by the activator domain of the fusion protein.
  • activity is assessed by detection of increased phosphorylated ErbB-3 in cells of hearts treated with the bi-specific fusion compared to untreated or mock treated hearts.
  • Myocardial infarction is generated in C57BL/6 mice by ligation of the left coronary artery (LCA) following endotracheal intubation, ventilation and thoracotomy. Coronary occlusion is confirmed by acute inspection of color change of the left ventricle wall, and ST elevation on the electrocardiogram before chest closure. Sham-operated mice undergo the same surgical procedure without LCA ligation.
  • a composition comprising the representative bi-specific fusion protein of Example 1 is administered to a mouse following myocardial infarction, induced as described above. Administration is via intravenous injection (e.g., tail vein). Following administration, heart function is assessed as follows. Mice are anesthetized with chloral hydrate (400 mg/kg body weight, i.p.), and the right carotid artery is cannulated with a microtip pressure transducer (model SPR-671, Millar) for the measurements of left ventricular (LV) pressures and LV+ and ⁇ dP/dt in the closed-chest preparation. Measurements are compared to those obtained from untreated control mice to confirm that treatment with the bi-specific fusion protein affects heart function. A significant improvement is observed in heart function as assessed using at least one of these measurements.

Abstract

Bi-specific fusion proteins with therapeutic uses are provided, as well as pharmaceutical compositions comprising such fusion proteins, and methods for using such fusion proteins to repair damaged tissue. The bi-specific fusion proteins generally comprise: (a) a targeting polypeptide domain that binds to an ischemia-associated molecule; and (b) an activator domain that that detectably modulates the activity of a cellular network.

Description

RELATED APPLICATIONS
This application is a divisional of U.S. application Ser. No. 14/187,728, filed Feb. 24, 2014, which is a divisional application of U.S. application Ser. No. 13/112,907, filed May 20, 2011, now U.S. Pat. No. 8,691,771, which claims the benefit of and priority to U.S. Provisional Application Ser. No. 61/347,040, filed May 21, 2010, the entire content of each of which is herein incorporated by reference in their entirety.
TECHNICAL FIELD
The present invention relates generally to fusion proteins that have therapeutic uses, and more specifically to bi-specific fusion proteins, pharmaceutical compositions comprising such fusion proteins, and methods for using such fusion proteins to repair damaged tissue.
BACKGROUND
Myocardial infarction, commonly known as a heart attack, occurs when coronary artery obstruction cuts off the blood supply to part of the heart. The resulting lack of oxygen causes irreversible tissue damage (necrosis and apoptosis), due to the inability of the heart to sufficiently activate endogenous regeneration programs and self-repair. Such tissue damage is a leading cause of congestive heart failure, a condition in which the heart is no longer capable of effectively pumping blood. In the United States, there are more than a million heart attacks every year, and nearly 5 million people are afflicted with congestive heart failure.
There are no effective treatments for regenerating damaged cardiac tissue. Current therapies for congestive heart failure focus on preventing arrhythmia, progression of arteriosclerosis and recurrent myocardial infarction, but do not address the underlying tissue damage. More than half of patients diagnosed with congestive heart failure die within five years of diagnosis.
Stem cell therapy is a potential new strategy for cardiac repair. In the laboratory, it is possible to generate cardiac muscle cells from stem cells. This suggests that stems cells could be used to repair damaged tissue such as cardiac tissue in a patient; however, no therapeutic treatments based on such an approach are presently available. One difficulty that has been encountered in stem cell therapy is that of targeting sufficient numbers of stem cells to the damaged tissue to result in clinically significant repair.
There is, thus, a need in the art for methods for repairing or regenerating damaged tissues, including cardiac tissue, and for improving the targeting of cells such as stem cells to facilitate tissue repair. The present invention fulfills these needs, and provides other related advantages.
SUMMARY OF THE INVENTION
The present invention provides bi-specific fusion proteins, nucleic acid molecules encoding bi-specific fusion proteins and therapeutic methods that employ such bi-specific fusion proteins.
In certain aspects, the present invention provides bi-specific fusion proteins that comprise: (a) a targeting domain having a binding specificity to an ischemia-associated molecule; and (b) an activator domain having a binding specificity to a growth factor receptor or cytokine receptor, wherein upon exposure of the activator domain to the growth factor receptor or cytokine receptor, the activator domain binds the growth factor receptor or cytokine receptor so as to modulate regeneration of a cardiac tissue.
In some embodiments, the bi-specific protein comprises: (a) a targeting polypeptide domain that binds to an ischemia-associated molecule with a Kd (i.e., said binding exhibits a Kd) ranging from 10−6 to 10−12 M or better; and (b) an activator domain that that detectably modulates the activity of a cellular network (e.g., detectably modulates activation of a growth factor receptor or cytokine receptor). In certain embodiments, the targeting polypeptide domain binds to the ischemia-associated molecule with a Kd ranging from 10−7 to 10−12 M or better, or ranging from 10−8 to 10−12 M or better. In further embodiments, the Kd is determined using a biosensor, e.g., by surface plasmon resonance or resonant mirror analysis.
In addition to components (a) and (b), above, certain bi-specific fusion proteins provided herein further comprise: (c) a polypeptide linker wherein the polypeptide linker extends the half life of the bi-specific fusion protein. In some embodiments, the targeting domain is at the N-terminus or at the C-terminus of the activator domain. In other embodiments, the polypeptide linkers at the N-terminus or at the C-terminus of the targeting domain. In some embodiments, the targeting domain is at the amino terminus of the fusion protein and the activator domain is at the carboxy terminus of the fusion protein. Yet in other embodiments, the targeting domain is at the carboxy terminus of the fusion protein and the activator domain is at the amino terminus of the fusion protein. In some embodiments, the polypeptide linker has two termini, an N-terminus and a C-terminus, that is joined at one terminus via a peptide bond to the targeting polypeptide domain and is joined at the other terminus via a peptide bond to the activator domain. In certain such embodiments, the targeting peptide is linked to the N-terminus of the linker and the activator domain is linked to the C-terminus of the linker. In other such embodiments, the targeting peptide is linked to the C-terminus of the linker and the activator domain is linked to the N-terminus of the linker. In certain embodiments, the linker is non-immunogenic in humans (e.g., a human serum protein or derivative thereof). Representative such linkers comprise at least 100 consecutive amino acids that are at least 80% identical to a serum albumin amino acid sequence, such as a human alpha-fetoprotein sequence. In certain embodiments, the linker comprises or has an amino acid sequence recited in any one of SEQ ID NOs: 10-29.
In some embodiments, the bi-specific fusion protein comprises (a) a targeting domain having a binding specificity to an ischemia-associated molecule; (b) an activator domain that detectably modulates activation of a receptor; and (c) a polypeptide linker, wherein the polypeptide linker extends the half life of the bi-specific fusion protein.
In some embodiments, the bi-specific fusion protein comprises (a) a targeting domain having a binding specificity to a target molecule; (b) an activator domain having a binding specificity to a receptor, wherein upon exposure of the activator domain to the receptor, the activator domain binds the receptor so as to modulate activation of the receptor; and (c) a polypeptide linker, wherein the polypeptide linker extends the half life of the bi-specific fusion protein.
In some embodiments, the bi-specific protein comprises (a) a targeting domain having a binding specificity to a tissue-associated molecule; and (b) an activator domain having a binding specificity to a molecule associated with the surface of a cell in the tissue, wherein upon exposure of the activator domain to surface-associated molecule, the activator domain binds the membrane-associated molecule so as to modulate regeneration of the tissue, wherein the targeting domain and the activator domain are linked via a linker, and wherein the linker extends the half life of the bi-specific fusion protein.
In some embodiments, the bi-specific fusion protein comprises (a) a targeting domain having a binding specificity to a target molecule; (b) an activator domain having a binding specificity to a receptor, wherein upon exposure of the activator domain to the receptor, the activator domain binds the receptor so as to modulate tissue regeneration; and (c) a polypeptide linker, wherein the polypeptide linker extends the half life of the bi-specific fusion protein.
In some embodiments, the targeting domain binds to the molecule with a dissociation constant Kd ranging from 10−6 M to 10−12 M. In some embodiments, the targeting domain binds to a molecule selected from the group of mysosin, cardiac myosin, DNA, phosphatidylserine, collagen, or extracellular matrix proteins. For example, the targeting domain can be selected from the group of annexin, anti-myosin antibody, anti-DNA scFv, variants thereof, fragments thereof, and combinations thereof. In some embodiments, the scFv antibody has a sequence recited in any one of SEQ ID NOs: 1, 2, or 30. In some embodiments, annexin has a sequence recited in SEQ ID NO: 31.
In some embodiments, the activator domain binds specifically to a growth factor receptor or cytokine receptor. For example, the activator domain is selected from the group consisting of hepatocyte growth factor, vascular endothelial growth factor, fibroblast growth factor, neuregulin/heregulin, variant thereof, and portion thereof.
In other embodiments, the bi-specific fusion proteins comprises (a) a leader polypeptide that comprises a sequence recited in SEQ ID NO:41 or 42; (b) a targeting polypeptide domain that binds to an ischemia-associated molecule, said binding exhibiting a Kd ranging from 10−6 to 10−12 M or better; (c) a short connector polypeptide that comprises the sequence -Gly-Ala- or -Ala-Ser-; (d) a HSA polypeptide that comprises a sequence recited in any one of SEQ ID NOs:10, 12, 14-29 and 45); (e) a short connector polypeptide that comprises the sequence -Leu-Gln- or -Thr-Gly-; (f) an activator domain that that detectably modulates the activity of a cellular network; and (g) a hexahistidine-comprising polypeptide.
It will be apparent that the above components may be present in the bi-specific fusion protein in the order recited or in a different order (e.g., the locations of the targeting polypeptide domain and activator domain may be switched). Within certain such bi-specific fusion proteins, the targeting polypeptide domain comprises a sequence recited in SEQ ID NO:1, 2, 30 or 31; the HSA polypeptide comprises the sequence recited in SEQ ID NO:45; the activator domain comprises a sequence recited in any one of SEQ ID NOs: 32-40; and the hexahistidine-comprising polypeptide has a sequence recited in SEQ ID NO:43 or 44.
In certain embodiments of the bi-specific fusion proteins described above, the ischemia-associated molecule is a DNA molecule, myosin (e.g., a myosin subtype such as cardiomyosin) or phosphatidyl serine.
In certain embodiments of the bi-specific fusion proteins described above, the targeting polypeptide comprises an antibody variable region. In certain such embodiments, the targeting polypeptide comprises a scFv antibody. Representative such scFv antibodies comprise or have a sequence recited in SEQ ID NO:1 or SEQ ID NO:2.
In certain embodiments of the bi-specific fusion proteins described above, the activator domain is a growth factor polypeptide. Within certain such embodiments, the growth factor polypeptide binds to a receptor for IGF or HGF (e.g., the growth factor polypeptide comprises or has an amino acid sequence recited in any one of SEQ ID NOs:3-9).
The bi-specific binding agents provided herein are not necessarily limited to two binding specificities. In certain embodiments, in addition to the targeting domain, the bi-specific fusion protein comprises two or more activator domains that are linked directly or indirectly via peptide bonds and are selected from growth factor polypeptides and cytokine polypeptides.
In other aspects, the present invention provides pharmaceutical compositions, comprising a bi-specific fusion protein as described above in combination with a physiologically acceptable carrier.
Within still further aspects, methods are provided for treating pathological tissue damage in a patient, comprising administering a pharmaceutical composition to a patient suffering from pathological tissue damage, and thereby decreasing pathological tissue damage in the patient. In certain embodiments, the pathological tissue damage is heart tissue damage associated with myocardial infarction. In other embodiments, the pathological tissue damage is kidney tissue damage.
In some embodiments, methods are provided for promoting tissue regeneration in a patient. The methods comprise (a) providing a bi-specific fusion protein comprising (i) a targeting domain having a binding specificity to an ischemia-associated molecule; and (ii) an activator domain having a binding specificity to growth factor receptor or cytokine receptor; and (b) administering in a patient in need thereof a therapeutically effective amount of the bi-specific fusion protein whereby the targeting domain specifically binds to the ischemia-associated molecule thereby targeting the bi-specific fusion protein to a tissue and whereby upon exposure of the activator domain to the growth factor receptor or cytokine receptor, the activator domain specifically activates the growth factor receptor or cytokine receptor so as to promote tissue regeneration. In some embodiments, the methods comprise (a) providing a bi-specific fusion protein comprising (i) a targeting domain having a binding specificity to a target molecule; (ii) an activator domain having a binding specificity to a receptor; (iii) a polypeptide linker, wherein the polypeptide linker extends the half life of the bi-specific fusion protein; and (b) administering in a patient in need thereof a therapeutically effective amount of the bi-specific fusion protein whereby the targeting domain specifically binds to the target molecule thereby targeting the bi-specific fusion protein to a first cell and whereby upon exposure of the activator domain to the growth factor receptor, the activator domain specifically activates the receptor of a second cell of a tissue so as to promote tissue regeneration.
In certain embodiments, such methods further comprise the administration of stem cells to the patient. In some embodiments, upon administration of the bi-specific protein, the bi-specific protein prevents cell damage, increases survival, promotes cell growth, promotes motility of stem cells, recruits stem cells, promotes differentiation of stem cells.
Also provided herein are nucleic acid molecules encoding a bi-specific fusion protein as described above. In certain embodiments, the nucleic acid molecule is DNA, and the DNA further comprises transcriptional and translational regulatory sequences operably linked to the bi-specific fusion protein coding sequence, such that transcription and translation of the coding sequence occurs in at least one eukaryotic cell type.
These and other aspects of the present invention will become apparent upon reference to the following detailed description.
DESCRIPTION OF THE SEQUENCE LISTING
SEQ ID NO:1 is the amino acid sequence of the anti-DNA scFv SV-1.
SEQ ID NO:2 is the amino acid sequence of the anti-DNA scFv SV-22.
SEQ ID NO:3 is the amino acid sequence of a growth factor polypeptide corresponding to wild type human IGF-I (mature form).
SEQ ID NO:4 is the amino acid sequence of a growth factor polypeptide corresponding to human IGF-1 with D12A substitution.
SEQ ID NO:5 is the amino acid sequence of a growth factor polypeptide corresponding to human IGF-1 with E9A substitution.
SEQ ID NO:6 is the amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain N-K1 domain.
SEQ ID NO:7 is the amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain K1 domain.
SEQ ID NO:8 is the amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain N-K2 fusion.
SEQ ID NO:9 is the amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain K2 domain.
SEQ ID NO:10 is the amino acid sequence of a human serum albumin (HSA) linker with C34S and N503Q substitutions.
SEQ ID NO:11 is the nucleic acid sequence of an HSA linker with C34S and N503Q substitutions.
SEQ ID NO:12 is the amino acid sequence of HSA.
SEQ ID NO:13 is the nucleic acid sequence of HSA.
SEQ ID NO:14 is the amino acid sequence of an HSA linker with C34S and N503Q substitutions and a polypeptide connector.
SEQ ID NO:15 is the amino acid sequence of an HSA linker with C34S and N503Q substitutions and a polypeptide connector.
SEQ ID NO:16 is the amino acid sequence of an HSA linker with C34S and N503Q substitutions and a polypeptide connector.
SEQ ID NO:17 is the amino acid sequence of an HSA linker with C34S and N503Q substitutions and a polypeptide connector.
SEQ ID NO:18 is the amino acid sequence of an HSA linker with C34S and N503Q substitutions and a polypeptide connector.
SEQ ID NO:19 is the amino acid sequence of an HSA linker with a polypeptide connector.
SEQ ID NO:20 is the amino acid sequence of an HSA linker with a polypeptide connector.
SEQ ID NO:21 is the amino acid sequence of an HSA linker with a polypeptide connector.
SEQ ID NO:22 is the amino acid sequence of an HSA linker with a polypeptide connector.
SEQ ID NO:23 is the amino acid sequence of an HSA linker with a polypeptide connector.
SEQ ID NO:24 is the amino acid sequence of an HSA linker with C34S substitution, domain I.
SEQ ID NO:25 is the amino acid sequence of an HSA linker, domain II.
SEQ ID NO:26 is the amino acid sequence of an HSA linker with N503Q substitution, domain III.
SEQ ID NO:27 is the amino acid sequence of an HSA linker, domain I.
SEQ ID NO:28 is the amino acid sequence of an HSA linker, domain II.
SEQ ID NO:29 is the amino acid sequence of human alpha-fetoprotein.
SEQ ID NO:30 is the amino acid sequence of the anti-phosphatidylserine scFv PS4A7.
SEQ ID NO:31 is the amino acid sequence of human annexin V.
SEQ ID NO:32 is an amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain N-K1 domain.
SEQ ID NO:33 is an amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain K1 domain.
SEQ ID NO:34 is an amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain N-K2 domain.
SEQ ID NO:35 is an amino acid sequence of a growth factor polypeptide corresponding to human HGF alpha chain K2 domain.
SEQ ID NO:36 is an amino acid sequence of a growth factor polypeptide corresponding to human VEGF alpha monomer.
SEQ ID NO:37 is an amino acid sequence of a growth factor polypeptide corresponding to human VEGF alpha dimer.
SEQ ID NO:38 is an amino acid sequence of a growth factor polypeptide corresponding to human FGF2.
SEQ ID NO:39 is an amino acid sequence of a growth factor polypeptide corresponding to human NRG1 alpha, EGF-like domain.
SEQ ID NO:40 is an amino acid sequence of a growth factor polypeptide corresponding to human NRG1 alpha, full sequence.
SEQ ID NO:41 is an amino acid sequence of a bi-specific fusion protein leader polypeptide.
SEQ ID NO:42 is an amino acid sequence of a bi-specific fusion protein leader polypeptide.
SEQ ID NO:43 is an amino acid sequence of a C-terminal hexahistidine-comprising polypeptide.
SEQ ID NO:44 is an amino acid sequence of a C-terminal hexahistidine-comprising polypeptide.
SEQ ID NO:45 is an amino acid sequence of a HSA linker.
SEQ ID NO:46 is an amino acid sequence of a HSA linker with N-terminal and C-terminal short connector polypeptides.
SEQ ID NO:47 is an amino acid sequence of a HSA linker with N-terminal and C-terminal short connector polypeptides.
SEQ ID NO:48 is an amino acid sequence of a HSA linker with N-terminal and C-terminal short connector polypeptides.
SEQ ID NO:49 is an amino acid sequence of a HSA linker with N-terminal and C-terminal short connector polypeptides.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to bi-specific fusion proteins that comprise: (1) a targeting polypeptide domain that binds to an ischemia-associated molecule; and (2) an activator domain, such as a growth factor polypeptide or a cytokine polypeptide. In certain embodiments, the bi-specific fusion protein further comprises: (3) a polypeptide linker having two termini, an N-terminus and a C-terminus, that is joined at one terminus via a peptide bond to the targeting polypeptide domain and is joined at the other terminus via a peptide bond to the activator domain. Such bi-specific fusion proteins find use, for example, in recruiting cells that express one or more growth factor and/or cytokine (e.g., chemokine) receptors (e.g., stem cells, progenitor cells or immune system cells) to tissue following an ischemic event (e.g., to damaged cells). In vivo, the administration of such bi-specific fusion proteins may be used to facilitate repair or regeneration of damaged tissue.
The term “polypeptide” is used herein to refer to a molecule that consists of multiple amino acid residues linked by peptide bonds. This term carries no implication as to the number of amino acid residues so linked.
The term “bi-specific” as used herein, refers to the ability of the fusion protein to interact with two different ligands: an ischemia-associated molecule (bound by the targeting polypeptide domain) and a receptor for the activator domain. The binding properties of the targeting polypeptide domain and the activator domain are discussed in more detail below.
An “ischemia-associated molecule” is any molecule that is detected at a level that is significantly higher (e.g., at least 2-fold higher) following ischemia or hypoxia. Any suitable binding assay may be used to identify ischemia-associated molecules, including those provided herein. The increased level of molecule that is detected may be the result of upregulation or decreased turnover, or may be due to increased accessibility (e.g., resulting from cell damage). In certain embodiments, the ischemia-associated molecule is detected in a cell of post-ischemic tissue at a significantly higher level (e.g., at least 2-fold higher) than in a cell of the same tissue that has not undergone an ischemic event (i.e., the molecule is specific to or enriched in the post-ischemic tissue). In further embodiments, the ischemia-associated molecule is associated with cell damage (i.e., the molecule is detected at a significantly higher level in cells that are damaged than in undamaged cells of the same type).
Certain ischemia-associated molecules are enriched (2 fold or higher) in the heart after an ischemic event (or in a model system that is used to mimic ischemia in the heart). Such molecules include, for example, molecules that are exposed on myocytes or other cardiac cells that undergo necrosis (such as DNA) or apoptosis (e.g., phosphatidylserine) or molecules that are enriched in scarred heart tissue, such as collagen (collagen I, III), myosin (including the cell type-specific subtypes thereof), or other extracellular matrix proteins that are enriched in post ischemic hearts. Such molecules can be identified on the basis of enrichment following ischemia-reperfusion in vivo or in simulated ischemia-reperfusion in vitro, or following exposure to conditions such as hypoxia, decreased ATP, increased reactive oxygen species (ROS) or nitric oxide synthase (NOS) production, or serum starvation of cells cultured in vitro.
The Targeting Polypeptide Domain
Binding to the ischemia-associated molecule is mediated by the targeting polypeptide domain. This domain may be any polypeptide sequence that serves this function; in preferred embodiments, the targeting polypeptide domain comprises one or more antibody variable regions.
As used herein, an “antibody” is a protein consisting of one or more polypeptides substantially encoded by immunoglobulin genes. A typical antibody is a tetramer that is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kD) and one “heavy” chain (about 50-70 kD). “VL” and VH” refer to these light and heavy chains respectively. An “antibody variable region” is an N-terminal region of an antibody variable chain (VL or VH) comprising amino acid residues that are primarily responsible for antigen recognition. Those of ordinary skill in the art are readily able to identify an antibody variable region and to determine the minimum size needed to confer antigen recognition. Typically, an antibody variable region comprises at least 70 amino acid residues, and more commonly at least 100 amino acid residues. A polypeptide that comprises an antibody variable region may (but need not) further comprise other light and/or heavy chain sequences, and may (but need not) further comprise sequences that are not antibody-derived. It will be apparent that the sequence of an antibody variable region may be naturally-occurring, or may be modified using standard techniques, provided that the function (antigen recognition) is retained. Certain polypeptides that comprise an antibody variable region are single chain antibodies (antibodies that exist as a single polypeptide chain), more preferably single chain Fv antibodies (scFv) in which a variable heavy chain region and a variable light chain region are joined together (directly or through a peptide linker) to form a continuous polypeptide. The scFv antibody may be chemically synthesized or may be expressed from a nucleic acid including VH- and VL-encoding sequences either joined directly or joined by a peptide-encoding linker.
“Binding” indicates that an antibody exhibits substantial affinity for a specific antigen (e.g., an ischemia-associated molecule) and is said to occur when the fusion protein (or the targeting polypeptide domain thereof) has a substantial affinity for the target antigen and is selective in that it does not exhibit significant cross-reactivity with other antigens. Preferred substantial binding includes binding with a dissociation constant (Kd) of 10−6, 10−7, 10−8, 10−9, 10−10, 10−11, 10−12 M or better. The Kd of an antibody-antigen interaction indicates the concentration of antibody (expressed as molarity) at which 50% of antibody and antigen molecules are bound together at thermodynamic equilibrium. Thus, at a suitable fixed antigen concentration, 50% of a higher (i.e., stronger) affinity antibody will bind antigen molecules at a lower antibody concentration than would be required to achieve the same percent binding with a lower affinity antibody. Kd is also the a ratio of the kinetic on and off rates (kon and koff); i.e., Kd=koff/kon. Thus, a lower Kd value indicates a higher (stronger) affinity. As used herein, “better” affinities are stronger affinities, and are identified by dissociation constants of lower numeric value than their comparators, with a Kd of 10−10M being of lower numeric value and therefore representing a better affinity than a Kd of 10−9M. Affinities better (i.e., with a lower Kd value and therefore stronger) than 10−7M, preferably better than 10−8M, are generally preferred. Values intermediate to those set forth herein are also contemplated, and preferred binding affinity can be indicated as a range of dissociation constants, for example preferred binding affinities for antibodies disclosed herein are represented by Kd values ranging from 10−6 to 10−12 M (i.e., micromolar to picomolar), preferably 10−7 to 10−12 M, more preferably 10−8 to 10−12 M or better. An antibody that “does not exhibit significant cross-reactivity” is one that will not appreciably bind to an off-target antigen. For example, in one embodiment, an antibody that specifically and selectively binds to Annexin V will exhibit at least a two, and preferably three, or four or more orders of magnitude better binding affinity (i.e., binding exhibiting a two, three, or four or more orders of magnitude lower Kd value) for Annexin V than for Annexin molecules other than Annexin V or for non-Annexin proteins or peptides. Binding affinity and selectivity can be determined using any art-recognized methods for determining such characteristics, including, for example, using Scatchard analysis and/or competitive (competition) binding assays.
Binding may be assessed, and Kd values determined, using any of a variety of techniques that are well known in the art. For example, binding to an ischemia-associated DNA molecule is commonly assessed by coating an appropriate solid support (e.g., beads, ELISA plate or BIACORE chip) with target DNA fragments. For a targeting polypeptide domain that binds to any sequence of DNA, DNA fragments (single or double-stranded) of 10 base pairs or larger are immobilized on the solid substrate. For a targeting polypeptide domain that binds to a specific sequence or DNA complex (e.g., DNA-histone complex) the appropriate corresponding target is immobilized. Prior to adding the ischemia-associated molecule, non-specific binding sites for protein are blocked with BSA, milk, or any other appropriate blocker. Uncoated wells or wells coated with a non-target molecule serve as specificity controls. Increasing concentrations of the bi-specific fusion protein (or targeting polypeptide domain) are incubated with target-coated substrate or control substrate. A fusion protein or domain that does not bind to the target is also tested as a specificity control. Target specific, dose-dependent binding of the bi-specific fusion protein (or targeting polypeptide domain) is then assessed by measuring the amount of bi-specific fusion protein (or targeting polypeptide domain) binding to target versus controls as a function of increasing dose using standard protocols corresponding to the solid support and binding technology being used. Representative such protocols include those described in Wassaf et al., Anal. Biochem. 351(2):241-53 (2006); Epub 2006 Feb. 10 (BIACORE); and Murray and Brown, J. Immunol. Methods. 127(1):25-8 (1990) (ELISA). In addition, studies that vary the amount of immobilized target molecule or that include increasing levels of soluble target molecule as a competitor may also be performed to monitor binding and specificity.
The binding affinity and kinetic on and off rates for binding to the target molecule are measured using standard techniques and compared to other negative control molecules (e.g., fusion protein with irrelevant targeting polypeptide or fusion protein lacking a targeting polypeptide) and positive control molecules (e.g., parental antibody that targets the ischemia-associated molecule, or other antibodies or antibody fragments that are known to bind to the ischemia-associated molecule).
In certain embodiments, the Kd is determined using a biosensor (e.g., by surface Plasmon resonance (BIAcore) or resonant mirror analysis (IAsys)). Such determinations may be performed as described by Hefta et al., Measuring Affinity Using Biosensors, in “Antibody Engineering: A Practical Approach,” McCafferty et al. (eds), pp. 99-116 (Oxford University Press, 1996), and references cited therein. Briefly, kinetic on and off rates (kon, and koff) are determined using a sensor chip to which the ischemia-associated molecule has been coupled. To evaluate association (kon), solutions of different concentrations of bi-specific fusion protein (or targeting polypeptide domain) flow across the chip while binding is monitored using mass sensitive detection. Using the BIAcore system (GE Healthcare; Piscataway, N.J.), kon, is the slope of the plot of dR/dt versus R, where R is the signal observed. Following binding, dissociation is observed by passing a buffer solution across the chip, and koff is determined in an analogous fashion. Kd is then calculated using the equation:
K d =k off /k on
In the context of the present invention, a bi-specific fusion protein binds to the ischemia-associated molecule if it binds with a Kd of less than 10−8 M, preferably less than 10−7 M, 10−8 M, 10−9 M or 10−10 M. In addition, the binding of the bi-specific fusion protein to the ischemia-associated molecule in this assay is significantly higher (e.g., at least 2-, 10- or 100-fold higher) than binding of the bi-specific fusion protein to negative controls. Preferably, binding to the immobilized target can also be competed using excess soluble target.
As noted above, certain ischemia-associated molecules are specific to (or enriched in) damaged cells. Binding to damaged cells is conveniently demonstrated in vitro using cultured cells that are exposed to conditions that induce necrosis or apoptosis. For example, necrosis can be induced in cultured cardiomyocytes by simulated ischemia/reperfusion, and monitored using a LDH release assay, or trypan blue assay followed by subtraction of the number of cells undergoing apoptosis, essentially as described in Shan et al., Am. J. Physiol. Cell. Physiol. 294:833-841 (2008). This assay quantitates the total dead cells and the difference between the total and the number of apoptotic cells is attributed to necrosis, as discussed in more detail below. Conditions that induce apoptosis include exposure to H2O2, and apoptosis can be monitored using any of a variety of techniques known in the art including, for example, annexin V (SEQ ID No. 31) binding cleavage of target peptide sequences by known caspases that are activated by apoptosis, or DNA laddering (measured by TUNEL assay, essentially as described in Kuramochi, J. Biol. Chem. 279(49): 51141-47 (2004)). Binding to the cells undergoing necrosis or apoptosis may be assessed by adding fluorescently labeled bi-specific fusion protein (or targeting polypeptide domain) or appropriate control proteins to cells following the induction of apoptosis or necrosis. After incubation of the proteins with the cells for times ranging from a few minutes to one day, the cells are washed and then the cell-bound fluorescence is measured using immunofluorescence, flow cytometry, or similar techniques. Alternatively, other methods of detecting the bound bi-specific fusion protein (or targeting polypeptide domain) may be used, including radiolabeling or using enzymes conjugated to the bi-specific fusion protein (or targeting polypeptide domain) or to antibodies that bind to the fusion protein (or targeting polypeptide domain), which is common practice in ELISA protocols. The bi-specific fusion protein (or targeting polypeptide domain) binds to target cells if significantly higher (e.g., 2-fold higher) binding to cells following ischemia (e.g., cells undergoing necrosis or apoptosis) is detected, as compared to cells that have not experienced an ischemic event (e.g., cells not undergoing apoptosis or necrosis).
In vivo targeting may be demonstrated by inducing ischemia in an animal model and comparing the level of administered bi-specific fusion protein (or targeting polypeptide domain) in a target tissue before and after ischemia. In vivo targeting to damaged cells may be demonstrated by inducing tissue damage in an animal model, administering the bi-specific fusion protein (or targeting polypeptide domain), and comparing the level of bi-specific fusion protein (or targeting polypeptide domain) in damaged versus undamaged cells. In one embodiment, the bi-specific fusion proteins are designed to target areas of tissue damage following ischemia-reperfusion injury. In such a case, demonstration of in vivo targeting may be accomplished by inducing tissue damage, preferably by a method that causes ischemia followed by re-establishment of blood supply. Numerous methods are available to do this in different tissues. For example, blood flow to the hindlimb of the mouse can be transiently blocked with a simple tourniquet. Alternatively, temporary clamp on the artery leading into the kidney can be employed. Ischemia-reperfusion injury can be induced in the heart through temporary blockage of the coronary artery as demonstrated in mice, rats, dogs, and pigs. Representative methods for inducing tissue damage in an animal model are summarized in Table 1.
TABLE 1
Representative Methods used to Induce Ischemia-Reperfusion Damage
Organ or
tissue Methods used to induce damage Reference
Heart Mouse: left anterior descending artery Dumont et al., Circulation
clamped for up to 30 minutes followed 102(13): 1564-8 (2000)
by reperfusion Davis, Proc. Natl. Acad. Sci. USA
Rat: coronary artery ligation 23: 103(21): 8155-60 (2006)
Kidney Mouse: Renal artery clamped with Chen et al., FASEB J. 4(12): 3033-39
pediatric suture for 1-6 hrs (1990)
Liver Dog: The hepatic pedicle and hepatic Miranda et al., Braz. J. Med. Biol. Res.
artery (close to the celiac artery) were 40(6): 857-65 (2007)
cross-clamped with vascular clamps. Kobayashi et al., World J.
Pig: Details in reference Gastroenterol.13(25): 3487-92 (2007)
Hindlimb Zbinden et al., Am. J. Physiol. Heart
Circ. Physiol. 292: H1891-H1897
(2007)
Animal models for ischemia-reperfusion injury are further detailed in the following references:
  • Greenberg et al., Chapter 7. Mouse models of ischemic angiogenesis and ischemia-reperfusion injury. Methods Enzymol. 444:159-74 (2008).
  • Chimenti et al., Myocardial infarction: animal models. Methods Mol. Med. 98:217-26 (2004).
  • Black S C, In vivo models of myocardial ischemia and reperfusion injury: application to drug discovery and evaluation. J. Pharmacol. Toxicol. Methods 43(2):153-67 (2000).
The specificity of targeting can be established by comparing the bi-specific fusion protein (or targeting polypeptide domain) deposition in the clamped versus unclamped kidney as shown in Chen et al., FASEB J. 4(12): 3033-39 (1990), or in the treated versus untreated hindlimb as shown in Zbinden et al., Am. J. Physiol. Heart Circ. Physiol. 292: H1891-H1897 (2007), using radiolabeled bi-specific fusion protein (or targeting polypeptide domain). Alternatively, bi-specific fusion protein (or targeting polypeptide domain) can be detected in homogenized tissue using ELISA, or can be imaged in real time using bi-specific fusion protein (or targeting polypeptide domain) labeled with the appropriate metal for imaging (e.g., Tc99, Y or Gd). Specific deposition in the damaged area of the heart can be measured as described in Dumont et al., Circulation 102(13):1564-8 (2000). Representative methods for demonstrating targeting of proteins to damaged tissue are shown in Table 2.
TABLE 2
Demonstration of Targeting to Damaged Tissue
Damaged organ
or tissue targeted Methods used to demonstrate targeted delivery Reference
Heart Humans: Tc99 labeling of Annexin V followed by Hofstra et al., The
imaging in humans using SPECT in patients with Lancet 356 (9225):
myocardial infarction followed by reperfusion attempts 209-12 (2000)
via angioplasty or thrombolysis
Heart Mouse: Fluorescent labeling of Annexin V in murine Dumont et al.,
model of ischemia reperfusion with distribution in the Circulation
myocardium detected histologically 102(13): 1564-8
(2000)
Heart Humans: Tc99 labeling of Annexin V followed by Hofstra et al., The
imaging in humans using SPECT in patients Lancet 356 (9225):
undergoing cardiac transplant rejection 209-12 (2000)
Heart Mouse: Fluorescently-labeled growth factor imaged in Urbanek, Proc.
heart tissue using confocal microscopy Natl. Acad. Sci.
USA 102
(24): 8692-97
(2005)
Damaged kidney Radiographs of clamped versus unclamped kidney Chen et al., FASEB
targeted using Microautoradiographs to show localization to specific J. 4(12): 3033-9
radiolabeled cellular structures in the kidney (1990)
antibody to DNA Imaging of whole mouse using I131-labeled antibody
to DNA (versus labeled control)
Biodistribution of I125-labeled antibody to show
deposition in non-target tissues
As noted above, certain targeting polypeptide domains comprise a scFv antibody that binds to the ischemia-associated molecule. Representative such scFv antibodies comprise or have the sequences provided herein as SEQ ID NOs: 1, 2, and 30.
It will be apparent that functionally related antibodies may also, or alternatively, be used as a targeting polypeptide domain. Antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain complementarity determining regions (CDRs). For this reason, the amino acid sequences within CDRs are more diverse between individual antibodies than sequences outside of CDRs. Because CDR sequences are responsible for most antibody-antigen interactions, it is possible to generate modified antibodies that mimic the properties of an original antibody by combining CDR sequences from one antibody with framework sequences from a different antibody. Such framework sequences can be obtained from public DNA databases that include germline antibody gene sequences.
Thus, one or more CDRs of a targeting polypeptide domain sequence provided herein, can be used to create functionally related antibodies that retain the binding characteristics of the original targeting polypeptide domain. In one embodiment, one or more CDR regions selected from SEQ ID NOs: 1, 2, and 30, is combined recombinantly with known human framework regions and CDRs to create additional, recombinantly-engineered, targeting polypeptide domains. The heavy and light chain variable framework regions can be derived from the same or different antibody sequences. CDR regions are readily identified using alignments with known sequences in databases such as Vbase and IMGT. The resulting targeting polypeptide domains share one or more CDRs with the targeting polypeptide domains of SEQ ID NOs: 1, 2, and 30; in certain embodiments, the targeting polypeptide domain comprises at least one CDR of a sequence as recited in SEQ ID NO: 1, 2, or 30.
It is well known in the art that antibody heavy and light chain CDR3 domains play a particularly important role in the binding specificity/affinity of an antibody for an antigen. Accordingly, in certain embodiments, antibodies are generated that include the heavy and/or light chain CDR3s of the particular antibodies described herein. The antibodies can further include the heavy and/or light chain CDR1 and/or CDR2s of the antibodies disclosed herein.
The CDR 1, 2, and/or 3 regions of the engineered antibodies described above can comprise the exact amino acid sequence(s) as those disclosed herein. However, the ordinarily skilled artisan will appreciate that some deviation from the exact CDR sequences may be possible, particularly for CDR1 and CDR2 sequences, which can tolerate more variation than CDR3 sequences without altering epitope specificity (such deviations are, e.g., conservative amino acid substitutions). Accordingly, in another embodiment, the engineered antibody may be composed of one or more CDR1s and CDR2s that are, for example, 90%, 95%, 98%, 99% or 99.5% identical to the corresponding CDRs of an antibody named herein.
In another embodiment, one or more residues of a CDR may be altered to modify binding to achieve a more favored on-rate of binding. Using this strategy, an antibody having ultra high binding affinity (e.g., Kd=10−10 or less) can be achieved. Affinity maturation techniques, well known in the art, can be used to alter the CDR region(s) followed by screening of the resultant binding molecules for the desired change in binding. Accordingly, as CDR(s) are altered, changes in binding affinity as well as immunogenicity can be monitored and scored such that an antibody optimized for the best combined binding and low immunogenicity are achieved.
Modifications can also be made within one or more of the framework or joining regions of the heavy and/or the light chain variable regions of an antibody, so long as antigen binding affinity subsequent to these modifications is not substantially diminished.
The Activator Domain
The activator domain is any polypeptide that detectably modulates the activity of a cellular network; certain activator domains are growth factor polypeptides or cytokine polypeptides (e.g., a chemokine polypeptide). It will be apparent that such modulation may be an increase or a decrease in the activity of the cellular network. A growth factor polypeptide detectably modulates activation of a growth factor receptor (such as HGF or IGF receptor). Certain such polypeptides are wild-type hepatocyte growth factor (HGF) or HGF alpha chain (e.g., GENBANK accession number P14210), or derivatives thereof that retain at least 10% of wild-type biological activity, as determined by measuring activation of the corresponding growth factor receptor in appropriate target cells. Activation may be assessed, for example, by measuring phosphorylation of receptor kinase or downstream proteins, such as AKT, essentially as described by Nishi et al., Proc. Natl. Acad. Sci. USA 95:7018-7023 (1998). MTT and CTG assays known in the art may also be used. Representative growth factor polypeptides have a sequence as recited in SEQ ID NO:3-9 or 32-40, herein. As discussed above for the targeting polypeptide domain, activator domains that share one or more CDRs with the activator domains of SEQ ID NOs: 3-9 or 32-40 are also contemplated; CDRs may be identified and such activator domains may be constructed using well known techniques. Thus, in certain embodiments, the activator domain comprises at least one CDR of a sequence as recited in SEQ ID NO:3-9 or 32-40. Similarly, a cytokine polypeptide modulates activation of the corresponding cytokine receptor, as determined in the same fashion.
In certain embodiments, the activator domain is a growth factor polypeptide, which binds a growth factor receptor on a cell surface. Representative such growth factor receptors are receptors for epidermal growth factor (EGF), Neregulin/Heregulin (NRG), fibroblast growth factor (FGF), insulin-like growth factor (e.g., IGF-I), platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF) and isoforms thereof (e.g., VEGF-A or VEGF-C), teratocarcinoma-derived growth factor 1 (TDGF1), transforming growth factor alpha (TGF-α), transforming growth factor beta (TGF-β) and isoforms thereof (e.g., TGF-β1 or TGF-(β2), thrombopoietin (THPO) or periostin. Other such receptors include mast/stem cell growth factor receptor (SCFR), hepatocyte growth factor receptor (HGF), ErbB-3, ErbB-4, high affinity nerve growth factor receptor, BDNF/NT-3 growth factors receptor, NT-3 growth factor receptor, or vascular endothelial growth factor receptor 1 (VEGFR-I). Representative cytokine receptors include, for example, FL cytokine receptor, receptor for cytokine receptor common gamma chain, interleukin-10 receptor alpha chain, interleukin-10 receptor beta chain, interleukin-12 receptor beta-1 chain, interleukin-12 receptor beta-2 chain, interleukin-13 receptor alpha-1 chain, interleukin-13 receptor alpha-2 chain, interleukin-17 receptor; interleukin-17B receptor, interleukin 21 receptor precursor, interleukin-1 receptor type I, interleukin-1 receptor type II, interleukin-2 receptor alpha chain, interleukin-2 receptor beta chain, interleukin-3 receptor alpha chain, interleukin-4 receptor alpha chain, interleukin-5 receptor alpha chain, interleukin-6 receptor alpha chain, interleukin-6 receptor beta chain, interleukin-7 receptor alpha chain, high affinity interleukin-8 receptor A, high affinity interleukin-8 receptor B, interleukin-9 receptor, interleukin-18 receptor 1, interleukin-1 receptor-like 1 precursor, interleukin-1 receptor-like 2, toll-like receptor 1, toll-like receptor 2, toll-like receptor 5, CX3C chemokine receptor 1, C-X-C chemokine receptor type 3, C-X-C chemokine receptor type 4, C-X-C chemokine receptor type 5, C-X-C chemokine receptor type 6, C-C chemokine receptor type 1, C-C chemokine receptor type 2, C-C chemokine receptor type 3, C-C chemokine receptor type 4, C-C chemokine receptor type 6, C-C chemokine receptor type 7 precursor, C-C chemokine receptor type 8, C-C chemokine receptor type 9, C-C chemokine receptor type 10, C-C chemokine receptor type 11, chemokine receptor-like 2, and chemokine XC receptor. Still other activator domains are receptors for solute carrier organic anion transporter family, member 1A2 (SLCO1A2), sphingosine kinase 1 (SPHK1), secreted phosphoprotein 1 (SPP1), also called osteopontin (OPN), tumor protein 53 (TP53), troponin T type 1 (TNNT1), TSPY-like protein 2 (TSPYL2), visfatin, WAP four-disulfide core domain 1 (WFDC1), thymosin beta 4, wingless-type MMTV integration site family, member 11 (WNT11). Representative activator domains include, for example, resistin, stromal cell-derived factor-1 (SDF-1), signal-induced proliferation-associated gene 1 (SIPA1), and any of the other ligands listed above, as well as portions and derivatives of the foregoing that substantially retain the ability to bind to cognate receptors.
As an initial test, binding of a bi-specific fusion protein (or activator domain thereof) to the appropriate receptor may be assessed using techniques known in the art. In one representative assay, binding is demonstrated by coating an appropriate solid support with the recombinant ectodomain of the appropriate receptor. An ectodomain from a receptor not recognized by the activator domain of the bi-specific fusion protein is used as a specificity control. A support substrate that does not have any immobilized receptor is also used as a control. Similar to the methods described above for binding to the ischemia-associated molecule, specific, dose-dependent binding to receptor is demonstrated using standard protocols corresponding to the solid support and binding technology being used. In addition, studies that vary the amount of receptor or that include increasing levels of soluble target molecule as a competitor are also performed to monitor binding and specificity. Alternatively, the bi-specific fusion protein is immobilized to a support and the binding of the soluble ectodomain of the corresponding receptor(s) is used to demonstrate dose-dependent, specific binding.
The binding affinity and kinetic on and off rates for binding of the bi-specific fusion protein to the receptor(s) are also measured using standard techniques and compared to other negative control molecules (fusion protein with irrelevant control activator domain, fusion protein lacking an activator domain) and positive control molecules (recombinant wild-type receptor ligand, such as a growth factor or cytokine). The equilibrium and kinetic binding parameters of the bi-specific fusion protein are also compared to the same parameters measured for the un-fused wild-type ligand to determine whether fusion of the ligand to other molecules affects the normal binding of the ligand to its corresponding receptor. Such information may be used to determine the effective dose of the bi-specific fusion protein.
A bi-specific fusion protein binds to immobilized growth factor receptor or cytokine receptor with a significantly higher affinity (e.g., at least 100-fold) than that observed for negative controls. In addition, binding to the immobilized receptor can be competed using excess soluble polypeptide, soluble receptor, or antibodies that bind to polypeptide or receptor and block their interaction. Preferably, the bi-specific fusion protein binds to the growth factor or cytokine receptor with an affinity within 1000-fold of the native ligand binding to its receptor.
A bi-specific fusion protein (and its activator domain) further has the capacity to mediate cognate receptor activation. Such activity may be assessed, for example, using a cellular model of ischemia reperfusion, which uses cultured cardiomyocytes such as neonatal rat ventricular myocytes (NRVM) or cell lines. Simulated ischemia (SI) is generally initiated by metabolic inhibitors (deoxyglucose and dithionite) and metabolites (high potassium, lactate, low pH) or by hypoxia in an anaerobic chamber. Reperfusion is simulated by resuspension in an oxygenated buffer. An in vitro adult cardiomyocyte pellet model of ischemia has been developed that provides the two primary components of ischemia—hypoxia and metabolite accumulation—in the absence of any exogenous metabolic inhibitors or metabolites. Table 3 shows representative methods for demonstrating the ability of a bi-specific fusion protein to prevent damage of cardiomyocytes, promote growth, motility or differentiation of cardiac stem cells and/or promote repair of damaged tissue.
TABLE 3
Activity Assessment Methods
Aspect Assay Reference
Localization and Detection of activator domain in cell lysate by Davis, Proc Natl Acad
retention kinetics ELISA Sci USA 103(21): 8155-60
of activator Detection of activator domain in cells by (2006)
domain immunofluorescence (flow cytometry or Urbanek, Proc. Natl.
microscopic) Acad. Sci. USA 102 (24):
8692-97 (2005)
Signaling by Detection of phospho-akt or phosphor-ERK in Davis, Proc Natl Acad
activator domain cells by flow cytometry, Sci USA 103(21): 8155-60
immunofluorescence, ELISA, phospho- (2006)
labeling, or Western Urbanek, Proc. Natl.
Acad. Sci. USA 102 (24):
8692-97 (2005)
Protection of cells AnnexinV binding by immunofluorescence or
against apoptosis flow cytometry
following hypoxia Detection of caspase activity
or other cell TUNEL-assay (reduced number of TUNEL-
stressor positive cells)
DNA laddering
Cell viability
Enhancement of cardiomyocyte viability
following exposure to H2O2. Number of
rod-shaped cells
pPCR assessment of gene expression
Protection of cells Reduced necrotic area by H&E staining
against necrosis
Reduction in scar Reduction in number of fibroblastic cells in
formation infarct area
Reduction collagen deposition
Reduction in other matrix proteins associated
with scar formation
Migration of CSC Time dependent increase in c-kit+, sca-1+, Urbanek, Proc. Natl.
into the infarct MDR1+ cell numbers and numbers Acad. Sci. USA 102 (24):
area undergoing transition to small myocytes 8692-97 (2005)
Myocyte Frequency of distribution of myocyte sizes Urbanek, Proc. Natl.
mechanics and cell Peak shortening Acad. Sci. USA 102 (24):
fusion: Velocity of shortening and relengthening 8692-97 (2005)
Assessment of cell fusion (number of X
chromosomes)
Cardiac functional Comparison of MI-treated versus MI- Urbanek, Proc. Natl.
assessment untreated animals Acad. Sci. USA 102 (24):
LVEDP 8692-97 (2005)
LVDP
+dp/dT
LV Weight
Chamber Volume
Diastolic Wall Stress
Survival
Myocardial Composition of regenerated myocardium Urbanek, Proc. Natl.
regeneration Assessment of BrdU+ cells in infarct area in Acad. Sci. USA 102 (24):
treated versus untreated animals 8692-97 (2005)
Myosin+ cells in the infarct area in treated
versus untreated animals
Cardiac structural Infarct size Urbanek, Proc. Natl.
Fibrosis Acad. Sci. USA 102 (24):
Cardiomyocyte hypertrophy 8692-97 (2005)
Native growth factors and cytokines can be used as activator domains. It will be apparent, however, that portions of such native sequences and polypeptides having altered sequences may also be used, provided that such polypeptides retain the ability to activate the cognate receptor (e.g., using one of the assays discussed below, such polypeptides detectably activate the receptor, and preferably activate the receptor to a degree that is at least 1% (preferably at least 10%) of that observed for the native ligand. Certain activator domains that bind to growth factor receptors are provided herein in SEQ ID NOs:3-9 and 32-40. Activity of fusion proteins comprising such sequences is well known in the art (e.g., Hashino et al., J. Biochem. 119(4):604-609 (1996); Nishi et al., Proc. Natl. Acad. Sci. USA 95:7018-23 (1998)).
An activator domain for a particular application may be selected based on the desired therapeutic outcome. For example, an activator domain that comprises FGF2, VEGF alpha or a portion or derivative thereof that substantially retains the ability to bind to cognate receptor, may generally be used to increase angiogenesis. To increase survival and for stem cell differentiation (regenerative) purposes, activator domains that comprise IGF, HGF or NRG1 (or a portion or derivative thereof) may be used.
In some cases, it may be desirable to assess the activity of both the activator domain and the targeting polypeptide simultaneously. An ELISA may be conveniently used for this purpose.
The substrate of the targeting polypeptide (e.g., DNA) is adsorbed to the ELISA plate, which is then blocked with appropriate BSA containing buffers. The bi-specific fusion protein is then added, followed by addition of recombinant substrate for the activator domain (e.g., if the activator is a growth factor, then the substrate is recombinant cognate receptor or receptor fragment (ectodomain)). This substrate is either fluorescently labeled for detection or detected using a labeled antibody to a region of the receptor that does not significantly affect ligand binding.
The in vivo activity of the bi-specific fusion protein is generally assessed by detecting signaling changes in molecules that are regulated by the activator domain of the bi-specific fusion protein. This typically involves changes in cell surface receptor phosphorylation status or downstream mediators such as phospho-AKT or phospho-ERK as detected by flow cytometry, immunofluorescence, ELISA, phospho-labeling, or Western analysis of treated tissues. Other functional assessments include tests for the number of viable cells by staining and morphological identification, level of apoptosis by Annexin V binding (via immunofluorescence) or flow cytometry, detection of caspase activity, TUNEL-assay (reduced number of TUNEL-positive cells) or DNA laddering. In each case, a bi-specific fusion protein functions in vivo if it induces a significant (e.g., at least 2-fold) change in the level, functional activity or phosphorylation of the regulated molecule detected by the assay.
The repair of damaged tissue in a patient can be assessed using any clinically relevant standard. For example, repair of infracted tissue can be measured by quantitation of cell number, such as the number of myocytes, fibroblast, or amount of scarring, or with functional assays for output or structural aspects of heart function including, LVEDP, LVDP, +dp/dT, LV Weight, Chamber Volume, and Diastolic Wall Stress. Methods for such assessments are well known and amply described in the literature. In general, a bi-specific fusion protein is said to repair damaged tissue if it results in a significant (e.g., at least 2-fold) change in any such clinical assessment.
Polypeptide Linker
The targeting polypeptide domain and activator domain may be directly joined via a peptide bond. Alternatively, they may be joined via a polypeptide linker. It will be apparent that any such linker will have two termini, an N-terminus and a C-terminus. The linker is joined at one terminus via a peptide bond to the targeting polypeptide domain and is joined at the other terminus via a peptide bond to the activator domain. In certain embodiments, the linker is joined at the N-terminus to the C-terminus of the targeting polypeptide domain and at the C-terminus to the N-terminus of the activator domain. In other embodiments, the linker is joined at the C-terminus to the targeting polypeptide domain and at the N-terminus to the activator domain.
Preferably, the linker is non-immunogenic in humans. More preferably, the linker is a human serum protein or a derivative thereof that retains at least 50% sequence identity over a region that consists of at least 100 consecutive amino acids. In further embodiments, the linker comprises at least 100 consecutive amino acids that are at least 70%, 80%, 85%, 90% or 95% identical to a human serum albumin amino acid sequence or a human alpha-fetoprotein amino acid sequence. Representative such linkers include those recited in any one of SEQ ID NOs:10, 12, 14-29 and 45, which may be incorporated into a bi-specific fusion protein alone or using a short (e.g., from 2 to 20 amino acid residues) connector polypeptide at one or both ends. Suitable short connector polypeptides for use at the N-terminal end of the linker include, for example, dipeptides such as -Gly-Ala-(GA) and -Ala-Ser-(AS). Suitable short connector polypeptides for use at the C-terminal end of the linker include, for example, dipeptides such as -Leu-Gln-(LQ) and -Thr-Gly-(TG). SEQ ID NOs:46-49 recite the linker of SEQ ID NO:45 with representative connector dipeptides at both the N- and C-termini; it will be apparent, however, that such short connector polypeptides, if present, may be located at either one or both termini.
Certain preferred linkers provide a prolonged half-life of the bi-specific fusion protein, as compared to fusion protein without linker. The effect of a linker on half-life can be evaluated using an assay that determines stability under physiological conditions. For example, bi-specific fusion protein can be incubated at 37° C. in serum (e.g., human) for 120 hours, with samples removed at the start of incubation and every 24 hours thereafter. Binding assays as described above are then performed to detect the level of functional bi-specific fusion protein at each time point. This level is then compared to the level of bi-specific fusion protein constructed without linker (or using a different linker) to provide a half-life comparison.
Optional Elements and Representative Bi-Specific Fusion Proteins
It will be apparent that elements in addition to those described above may optionally be included in the bi-specific fusion proteins provided herein. Such elements may be present for a variety of purposes, including to facilitate expression, preparation or purification of the bi-specific fusion protein, or to perform targeting functions. For example, an N-terminal leader polypeptide may be present. Representative leader polypeptides comprise or have a sequence recited in SEQ ID NO:41 or 42. A bi-specific fusion protein may also, or alternatively, comprise a polyhistidine (e.g., hexahistidine) tag to facilitate purification. Such a tag comprises at least six histidine consecutive amino acid residues, and may be located at the C- or N-terminus. In certain embodiments, a hexahistidine tag is included at the C-terminus of the bi-specific fusion protein. Additional amino acid residues may also be present at the junction of the polyhistidine to the remainder of the bi-specific fusion protein. Certain bi-specific fusion proteins provided herein comprise a C-terminal polyhistidine-comprising polypeptide as recited in SEQ ID NO:43 or 44.
Certain bi-specific fusion proteins have a general structure that satisfies one of the following (shown from N-terminal to C-terminal, left to right):
Representative bi-specific fusion proteins comprise (from N-terminal to C-terminal):
(a) a leader polypeptide (e.g., comprising or having a sequence recited in SEQ ID NO:41 or 42);
(b) a targeting polypeptide domain (e.g., comprising or having a sequence recited in SEQ ID NO: 1, 2, 30 or 31);
(c) a short connector polypeptide (e.g., comprising or having the sequence Gly-Ala- or -Ala-Ser-);
(d) a HSA polypeptide (e.g., comprising or having a sequence recited in any one of SEQ ID NOs:10, 12, 14-29 and 45);
(e) a short connector polypeptide (e.g., comprising or having the sequence -Leu-Gln- or -Thr-Gly-);
(f) an activator domain (e.g. comprising or having a sequence recited in any one of SEQ ID NOs:3-9 and 32-40); and
(g) a polyhistidine-comprising polypeptide (e.g., a hexahistidine-comprising polypeptide, such as a polypeptide comprising or having a sequence recited in SEQ ID NO:43 or 44.
For example, certain such bi-specific fusion proteins comprise (N-terminal to C-terminal): a leader sequence as recited in SEQ ID NO:41 or 42; a targeting polypeptide domain as recited in SEQ ID NO:1, 2, 30 or 31; an HSA polypeptide having the sequence recited in SEQ ID NO:45; a -Gly-Ala or Ala-Ser- connector dipeptide; -Leu-Gln- or -Thr-Gly-; an activator domain having a sequence recited in any one of SEQ ID NOs: 32-40; and a hexahistidine-comprising polypeptide having a sequence recited in SEQ ID NO:43 or 44.
Other bi-specific fusion proteins comprise (from N-terminal to C-terminal):
(a) a leader polypeptide (e.g., comprising or having a sequence recited in SEQ ID NO:41 or 42);
(b) an activator domain (e.g., comprising or having a sequence recited in any one of SEQ ID NOs:3-9 and 32-40);
(c) a short connector polypeptide (e.g., comprising or having the sequence -Gly-Ala- or -Ala-Ser-);
(d) an HSA polypeptide (e.g., comprising or having a sequence recited in any one of SEQ ID NOs:10, 12, 14-29 and 45);
(e) a short connector polypeptide (e.g., comprising or having the sequence -Leu-Gln- or -Thr-Gly-);
(f) a targeting polypeptide domain (e.g., comprising or having a sequence recited in SEQ ID NO: 1, 2, 30 or 31); and
(g) a poly-histidine-comprising polypeptide (e.g., comprising or having as sequence recited in SEQ ID NO:43 or 44.
Still further bi-specific fusion proteins comprise (from N-terminal to C-terminal):
(a) a leader polypeptide (e.g., comprising or having a sequence recited in SEQ ID NO:41 or 42);
(b) an activator domain (e.g., comprising or having a sequence recited in any one of SEQ ID NOs:3-9 and 32-40);
(c) an HSA polypeptide that has a sequence recited in any one of SEQ ID NOs:46-49;
(d) a targeting polypeptide domain (e.g., comprising or having a sequence recited in SEQ ID NO: 1, 2, 30 or 31); and
(e) a poly-histidine-comprising polypeptide (e.g., comprising or having as sequence recited in SEQ ID NO:43 or 44.
Preparation of Bi-Specific Fusion Proteins
Bi-specific fusion proteins may be synthesized using standard techniques, including liquid- and solid-phase peptide synthesis and recombinant DNA techniques. For solid phase synthesis, the C-terminal amino acid of the sequence is attached to an insoluble support, and the remaining amino acids are added in sequence. For polypeptides longer than about 50 amino acids, shorter regions may be synthesized in this fashion and then condensed to form the longer polypeptide. Methods of forming peptide bonds by activation of a carboxyl terminal end (e.g., by the use of the coupling reagent N, N′-dicyclohexylcarbodiimide) are well known in the art.
For recombinant DNA techniques, DNA encoding the bi-specific fusion protein is prepared chemically or by isolating and ligating DNA encoding each portion of the fusion protein. The DNA coding for each segment of the bi-specific fusion protein may be isolated from known genes or synthesized de novo. Methods for direct chemical synthesis of DNA are well known in the art, and such syntheses are routinely performed using an automated synthesizer. Chemical synthesis produces a single stranded polynucleotide, which is converted into double stranded DNA by hybridization with a complementary sequence or using DNA polymerase. While chemical synthesis of DNA is generally limited to sequences that are shorter than the bi-specific fusion protein, it will be apparent that the full bi-specific fusion protein may be obtained by ligation of shorter sequences in frame. Alternatively, DNA sequences encoding the bi-specific fusion protein are prepared by cloning. Cloning techniques are well known in the art, and are amply described, for example, by standard references such as Sambrook et al., Molecular Cloning: A Laboratory Manual (3rd ed.), Cold Spring Harbor Laboratory Press (2001). Portions of the DNA may be ligated together in frame to generate the full length coding sequence.
Once the DNA encoding the bi-specific fusion protein is obtained, the DNA may be cloned into a vector for expression in a prokaryotic or eukaryotic host cell. Techniques for incorporating DNA into such vectors are well known to those of ordinary skill in the art. Within such an expression vector, the DNA encoding the bi-specific fusion protein is operably linked to the nucleotide sequences necessary for expression (e.g., a suitable promoter and, if necessary, a terminating signal). A promoter is a nucleotide sequence (typically located 5′ to the coding sequence) that directs the transcription of adjacently linked coding sequences. A terminating signal may be a stop codon to end translation and/or a transcription termination signal. Additional regulatory element(s) (e.g., enhancer elements) may also be present within an expression vector. Such a vector is preferably a plasmid or viral vector. Preferably, an expression vector further comprises a selectable marker, which confers resistance to a selection. This allows cells to stably integrate the vector into their chromosomes and grow to form foci, which in turn can be cloned and expanded into cell lines. A variety of selectable markers are known in the art, including, for example, genes that provide resistance to ampicillin, methotrexate, mycophenolic acid, the aminoglycoside G-418, hygromycin and puromycin. Those of ordinary skill in the art are knowledgeable in the numerous expression systems available for expression of proteins including E. coli, other bacterial hosts, yeast, and various higher eukaryotic cells such as the COS, CHO, HeLa and myeloma cell lines.
Host cells are transformed or transfected with the vector that comprises the DNA encoding the bi-specific fusion protein using standard methods. Expression in the host cell results in transcription of the DNA into the corresponding mRNA, followed by translation of the mRNA to generate the bi-specific fusion protein.
Once expressed, the bi-specific fusion protein can be purified according to standard procedures, including, for example, ammonium sulfate precipitation or affinity column chromatography. Substantially pure compositions of at least about 90 to 95% homogeneity are preferred, and 98 to 99% or more homogeneity is most preferred for pharmaceutical uses. Once purified, partially or to homogeneity as desired, if to be used therapeutically, the polypeptides should be substantially free of endotoxin.
Pharmaceutical Compositions
The present invention also provides pharmaceutical compositions comprising at least one bi-specific fusion protein as described herein, together with at least one physiologically acceptable carrier. Such compositions may be used for treating patients who are suffering from, or at risk for, tissue damage, in order to prevent tissue damage, or to repair or regenerate damaged tissue. Such patients include, for example, patients who have experienced myocardial infarction, kidney damage, and/or ischemic stroke). If desired, other active ingredients may also be included within the pharmaceutical composition, such as stem cells or other agents that facilitate repair of damaged tissue.
As used herein, the term “physiologically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term “carrier” refers to a diluent, adjuvant, excipient, or vehicle with which the bi-specific fusion protein is administered. Physiologically acceptable carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin (e.g., peanut oil, soybean oil, mineral oil, or sesame oil). Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include, for example, starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water and ethanol. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
Pharmaceutical compositions may be formulated for any appropriate manner of administration, including, for example, parenteral, intranasal, topical, oral, or local administration, such as by a transdermal means, for prophylactic and/or therapeutic treatment. These compositions can take any of a variety of well known forms that suit the mode of administration, such as solutions, suspensions, emulsions, tablets, pills, capsules, powders, aerosols and sustained-release formulations. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical modes of administration and carriers are described in “Remington: The Science and Practice of Pharmacy,” A. R. Gennaro, ed. Lippincott Williams & Wilkins, Philadelphia, Pa. (21st ed., 2005).
Commonly, the pharmaceutical compositions provided herein are administered parenterally (e.g., by intravenous, intramuscular, or subcutaneous injection), or by oral ingestion or topical application. For parenteral administration, the bi-specific fusion protein can either be suspended or dissolved in the carrier. A sterile aqueous carrier is generally preferred, such as water, buffered water, saline or phosphate-buffered saline. In addition, sterile, fixed oils may be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed, including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectible compositions. Pharmaceutically acceptable auxiliary substances may also be included to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, dispersing agents, suspending agents, wetting agents, detergents, preservatives, local anesthetics and buffering agents.
In one preferred embodiment, the pharmaceutical composition is formulated for intravenous administration to a patient (e.g., a human). Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a sealed (e.g., hermetically sealed) container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
Compositions intended for oral use may be presented as, for example, tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. Such compositions may further comprise one or more components such as sweetening agents flavoring agents, coloring agents and preserving agents. Tablets contain the active ingredient in admixture with physiologically acceptable excipients that are suitable for the manufacture of tablets. Such excipients include, for example, inert diluents, granulating and disintegrating agents, binding agents and lubricating agents. Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium. Aqueous suspensions comprise the active materials in admixture with one or more excipients suitable for the manufacture of aqueous suspensions. Such excipients include suspending agents and dispersing or wetting agents. Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil (e.g., arachis oil, olive oil, sesame oil or coconut oil) or in a mineral oil such as liquid paraffin. Pharmaceutical compositions may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil or a mineral oil or mixture thereof. Suitable emulsifying agents include, for example, naturally-occurring gums, naturally-occurring phosphatides and anhydrides.
Pharmaceutical compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered. Sterile aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of an aqueous pharmaceutical composition typically will be between 3 and 11, more preferably between 5 and 9 or between 6 and 8, and most preferably between 7 and 8, such as 7 to 7.5.
Bi-specific fusion proteins provided herein are generally present within a pharmaceutical composition at a concentration such that administration of a single dose to a patient delivers a therapeutically effective amount. A therapeutically effective amount is an amount that results in a discernible patient benefit, such as detectable repair or regeneration of damaged tissue or diminution of symptoms of tissue damage. Therapeutically effective amounts can be approximated from the amounts sufficient to achieve detectable tissue repair or regeneration in one or more animal models exemplified in Table 3. Nonetheless, it will be apparent that a variety of factors will affect the therapeutically effective amount, including the activity of the bi-specific fusion protein employed; the age, body weight, general health, sex and diet of the patient; the time and route of administration; the rate of excretion; any simultaneous treatment, such as a drug combination; and the type and severity of the tissue damage in the patient undergoing treatment. Optimal dosages may be established using routine testing, and procedures that are well known in the art. Dosages generally range from about 0.5 mg to about 400 mg of bi-specific fusion protein per dose (e.g., 10 mg, 50 mg, 100 mg, 200 mg, 300 mg, or 400 mg per dose). In general, compositions providing dosage levels ranging from about 0.1 g to about 100 g per kilogram of body weight per day are preferred. In certain embodiments, dosage unit forms contain between from about 10 g to about 100 g of bi-specific fusion protein.
Pharmaceutical compositions may be packaged for treating or preventing tissue damage (e.g., for treatment of myocardial infarction or kidney damage). Packaged pharmaceutical preparations include a container holding a therapeutically effective amount of at least one pharmaceutical composition as described herein and instructions (e.g., labeling) indicating that the contained composition is to be used for treating tissue damage (such as myocardial infarction or kidney damage) in a patient. Pharmaceutical compositions may be packaged in multiple single dose units, each containing a fixed amount of bi-specific fusion protein in a sealed package. Alternatively, the container may hold multiple doses of the pharmaceutical composition.
Methods of Treatment
The pharmaceutical compositions can be administered to a patient (preferably a mammal such as a cow, pig, horse, chicken, cat, dog, or more preferably a human) to treat pathological tissue damage in the patient. Within the context of the present invention, the term “treatment” encompasses both prophylactic and therapeutic administration. In prophylactic applications, a pharmaceutical composition as described herein is administered to a patient susceptible to or otherwise at risk for developing pathological tissue damage, in order to prevent, delay or reduce the severity of tissue damage. In therapeutic applications, treatment is performed in order to reduce the severity of the pathological tissue damage exist in the patient prior to treatment. Representative pathological tissue damage includes heart tissue damage (e.g., damage associated with myocardial infarction), kidney tissue damage and ischemic stroke.
Any of a variety of known delivery systems can be used to administer a bi-specific fusion protein including, for example, encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the bi-specific fusion protein, receptor-mediated, or a retroviral or other nucleic acid vector. The bi-specific fusion protein may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.), and may be administered together with other biologically active agents. Administration can be systemic or local. In addition, it may be desirable to introduce the bi-specific fusion protein into the central nervous system by any suitable route, including intraventricular and intrathecal injection; intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir. Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
In a specific embodiment, it may be desirable to administer the bsBAs of the invention locally to the area in need of treatment; this may be achieved by, for example, local infusion during surgery, topical application (e.g., in conjunction with a wound dressing after surgery), by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. In another embodiment, a vesicle, such as a liposome, can be used to deliver the bi-specific fusion protein. In yet another embodiment, the bi-specific fusion protein is delivered in a controlled release system; for example, such a controlled release system may be placed at or near the therapeutic target (e.g., an organ of the body that has experienced or is at risk for tissue damage). The use of such delivery systems is well known to those of ordinary skill in the art.
Without wishing to be bound by any particular theory, it is believe that the bi-specific fusion proteins provided herein are effective for treating pathological tissue damage at least in part due to their ability to recruit stem cells to the damaged tissue. In certain cases, sufficient stem cells may reside within the patient (e.g., resident cardiac stem cells). In certain embodiments, however, it may be beneficial to co-administer stem cells (e.g., bone marrow-derived autologous stem cells). Such stem cells may be administered before or after the bi-specific fusion protein, or may be administered simultaneously (either in the same pharmaceutical composition or in separate compositions).
As noted above, the optimal dose depends on certain factors known in the art, but generally ranges from about 0.5 mg to about 400 mg of bi-specific fusion protein per dose (e.g., 10 mg, 50 mg, 100 mg, 200 mg, 300 mg, or 400 mg per dose). A dose of bi-specific fusion protein (within a pharmaceutical composition as described above) can be administered therapeutically to a patient one or more times per hour, day, week, month, or year (e.g., 2, 4, 5, 6, 7, 8, 9, 10, 11, or 12 times per hour, day, week, month, or year). More commonly, a single dose per day or per week comprising an amount of bi-specific fusion protein ranging from about 0.1 g to about 100 g per kilogram of body weight is administered.
In other embodiments, a pharmaceutical composition comprising a bi-specific fusion protein may be administered to a patient in a dosage that ranges from about 0.5 mg per week to about 400 mg per week, about 1.0 mg per week to about 300 mg per week, about 5 mg per week to about 200 mg per week, about 10 mg per week to about 100 mg per week, about 20 mg per week to about 80 mg per week, about 100 mg per week to about 300 mg per week, or about 100 mg per week to about 200 mg per week. Alternatively, a pharmaceutical composition comprising a bi-specific fusion protein may be administered at a dose that ranges from about 0.5 mg every other day to about 100 mg every other day, about 5 mg every other day to about 75 mg every other day, about 10 mg every other day to about 50 mg every other day, or about 20 mg every other day to about 40 mg every other day. A pharmaceutical composition comprising a bi-specific fusion protein may alternatively be administered at a dose that ranges from about 0.5 mg three times per week to about 100 mg three times per week, about 5 mg three times per week to about 75 mg three times per week, about 10 mg three times per week to about 50 mg three times per week, or about 20 mg three times per week to about 40 mg three times per week.
In further embodiments of, a pharmaceutical composition comprising a bi-specific fusion protein is administered to a mammal (e.g., a human) continuously for 1, 2, 3, or 4 hours; 1, 2, 3, or 4 times a day; every other day or every third, fourth, fifth, or sixth day; 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times a week; biweekly; 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 times a month; bimonthly; 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times every six months; 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 times a year; or biannually. It will be apparent that a pharmaceutical composition comprising a bi-specific fusion protein may, but need not, be administered at different frequencies during a therapeutic regime.
The following Examples are offered by way of illustration and not by way of limitation. Unless otherwise specified, all reagents and solvents are of standard commercial grade and are used without further purification. Using routine modifications, the procedures provided in the following Examples may be varied by those of ordinary skill in the art to make and use other bi-specific fusion proteins and pharmaceutical compositions within the scope of the present invention.
EXAMPLES Example I Preparation of a Representative Bi-Specific Fusion Protein
A bi-specific fusion protein in which targeting polypeptide domain binds to DNA and the activator domain is NRG1 is prepared. The two domains are joined by a modified human serum albumin (HSA) linker. The NRG1 is recombinantly fused to the amino terminus of the HSA linker incorporating a short connector polypeptide and the anti-DNA scFv is recombinantly fused to the carboxy terminus of the modified HSA linker incorporating an additional short connector polypeptide. The modified HSA linker contains two amino acid substitutions. A cysteine residue at position 34 of native HSA is mutated to serine in order to reduce potential protein heterogeneity due to oxidation at this site. An asparagine residue at amino acid 503 of native HSA, which may be sensitive to deamidation, resulting in decreased pharmacologic half-life, is mutated to glutamine. The modified HSA linker confers an extended circulating half-life on the bi-specific fusion protein.
Example II In vitro Activity of a Bi-Specific Fusion Protein
The activity of both components of the representative bi-specific fusion protein prepared in Example 1 (in which the targeting polypeptide domain binds to DNA and the activator domain is NRG1) are tested using an ELISA designed to give activity only when both arms of the bi-specific fusion protein are bound to their substrates simultaneously. The ELISA is performed essentially as described in Stokes et al., J. Clin. Pathol. 35(5): 566-573 (1982) and Gripenberg et al., Scand. J. Immunol. 1:151-157 (1978). More specifically, 1 to 50 ng/ml solution of the bi-specific fusion protein in PBS is added to the wells of a plate pre-adsorbed with DNA (Anti-DS-DNA antibody ELISA kit (Alpha Diagnostic International, Dist by AutogenBioclear, UK) and incubated and washed according to manufacturer's directions until the step in which the detection antibody is added. At this stage, 100 μl of 1-50 ng/ml solution of Biotinylated goat anti-human NRG1-β1 (R&D Systems BAF377) (antibody to the ‘activator arm’) in PBS/1% BSA/0.05% Tween is added to all wells and incubated for 1 hr at room temperature, washed in PBS with 0.05% Tween-20. 100 μl of Streptavidin-HRP (1:200 dilutions of stock 2 μg/ml, (R&D Systems 890803)) diluted in PBS is added to each well and incubated 30 min at room temperature. After a final wash in PBS with 0.05% Tween-20, 100 μl of SuperSignal ELISA Pico Chemiluminescent Substrate (as per manufacturer's instructions, Pierce, cat #34077) is added and luminescence (representative of positive signal) is measured on Fusion Microplate reader (Packard) or similar instrument.
The amount of signal detected is significantly higher (at least 100-fold higher) in the wells with bi-specific fusion protein than in wells without DNA or negative controls that contain a dead arm (i.e., does not contain an activator domain or targeting polypeptide domain). In addition, the signal is seen to vary with the amount of bi-specific fusion protein added to the wells.
Example III In vivo Activity of a Bi-Specific Fusion Protein
The in vivo activity of the representative bi-specific fusion protein prepared in Example 1 is determined by detecting signaling changes in a molecule that is regulated by the activator domain of the fusion protein. For the activator domain in this fusion protein NRG1, activity is assessed by detection of increased phosphorylated ErbB-3 in cells of hearts treated with the bi-specific fusion compared to untreated or mock treated hearts. Myocardial infarction is generated in C57BL/6 mice by ligation of the left coronary artery (LCA) following endotracheal intubation, ventilation and thoracotomy. Coronary occlusion is confirmed by acute inspection of color change of the left ventricle wall, and ST elevation on the electrocardiogram before chest closure. Sham-operated mice undergo the same surgical procedure without LCA ligation.
Hearts from normal mice or those following induction of myocardial infarction, from both control and bi-specific fusion protein treated mice, are removed, fixed in 4% paraformaldehyde, embedded, sectioned and mounted as described in Dhein, Mohr and Delmar, Practical Methods in Cardiovascular Research, 2005, p. 473 (Springer, N.Y.). Phospho-ErbB3 antibody (Cell Signaling Technology; Beverly, Mass.) is used for detection of Phospho-ErbB3 by immunofluorescence. A 2-fold increase or more in phospho-ErbB3 levels in treated versus untreated hearts is observed and is indicative of functional activator. The increase is in either the number (number per field, or percentage of total) of cells exhibiting signal, the intensity of signal per cell, or both.
Example IV Tissue Damage Repair in Mice using a Bi-Specific Fusion Protein
A composition comprising the representative bi-specific fusion protein of Example 1 is administered to a mouse following myocardial infarction, induced as described above. Administration is via intravenous injection (e.g., tail vein). Following administration, heart function is assessed as follows. Mice are anesthetized with chloral hydrate (400 mg/kg body weight, i.p.), and the right carotid artery is cannulated with a microtip pressure transducer (model SPR-671, Millar) for the measurements of left ventricular (LV) pressures and LV+ and −dP/dt in the closed-chest preparation. Measurements are compared to those obtained from untreated control mice to confirm that treatment with the bi-specific fusion protein affects heart function. A significant improvement is observed in heart function as assessed using at least one of these measurements.
It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications of changes in light thereof are to be included within the spirit and purview of this application and scope of the appended claims. All publication, patents and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes.

Claims (7)

The invention claimed is:
1. A bi-specific fusion protein comprising:
(a) a targeting domain comprising Annexin V having the amino acid sequence recited in SEQ ID NO: 31;
(b) an activator domain comprising neuregulin/heregulin having the amino acid sequence recited in SEQ ID NO: 39 or SEP ID NO: 40; and
(c) a polypeptide linker comprising human serum albumin having the amino acid sequence recited in SEQ ID NO: 10.
2. The bi-specific fusion protein of claim 1, wherein the activator domain binds specifically to neuregulin/heregulin receptor.
3. The bi-specific fusion protein of claim 1, wherein the targeting domain binds to the phosphatidylserine with a dissociation constant Kd ranging from 10−6 M to 10−12 M.
4. The bi-specific fusion protein of claim 1, wherein the linker is a non-immunogenic polypeptide.
5. The bi-specific fusion protein of claim 1, wherein the linker extends a half-life of the bi-specific fusion protein.
6. A pharmaceutical composition comprising a physiologically acceptable carrier and a therapeutically effective amount of the bi-specific fusion protein of claim 1.
7. A method of treating a tissue damage in a subject, comprising administering the pharmaceutical composition of claim 6 to a patient in need thereof so as to modulate tissue regeneration.
US15/957,252 2010-05-21 2018-04-19 Bi-specific fusion proteins Active US10858450B2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/957,252 US10858450B2 (en) 2010-05-21 2018-04-19 Bi-specific fusion proteins
US17/078,978 US11673970B2 (en) 2010-05-21 2020-10-23 Bi-specific fusion proteins
US18/311,306 US20230322951A1 (en) 2010-05-21 2023-05-03 Bi-Specific Fusion Proteins

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US34704010P 2010-05-21 2010-05-21
US13/112,907 US8691771B2 (en) 2010-05-21 2011-05-20 Bi-specific fusion proteins for tissue repair
US14/187,728 US9982060B2 (en) 2010-05-21 2014-02-24 Bi-specific fusion proteins
US15/957,252 US10858450B2 (en) 2010-05-21 2018-04-19 Bi-specific fusion proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/187,728 Division US9982060B2 (en) 2010-05-21 2014-02-24 Bi-specific fusion proteins

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/078,978 Division US11673970B2 (en) 2010-05-21 2020-10-23 Bi-specific fusion proteins

Publications (2)

Publication Number Publication Date
US20180237540A1 US20180237540A1 (en) 2018-08-23
US10858450B2 true US10858450B2 (en) 2020-12-08

Family

ID=44972647

Family Applications (11)

Application Number Title Priority Date Filing Date
US13/068,808 Active US9238080B2 (en) 2010-05-21 2011-05-20 Bi-specific fusion proteins
US13/112,907 Active 2031-11-01 US8691771B2 (en) 2010-05-21 2011-05-20 Bi-specific fusion proteins for tissue repair
US14/187,728 Active 2031-10-04 US9982060B2 (en) 2010-05-21 2014-02-24 Bi-specific fusion proteins
US14/967,980 Active US9718892B2 (en) 2010-05-21 2015-12-14 Method of treating myocardial infarction by administering a bi-specific fusion protein
US15/618,478 Active US10407512B2 (en) 2010-05-21 2017-06-09 Bi-specific fusion proteins
US15/957,252 Active US10858450B2 (en) 2010-05-21 2018-04-19 Bi-specific fusion proteins
US16/524,451 Active US10988547B2 (en) 2010-05-21 2019-07-29 Bi-specific fusion proteins
US17/078,978 Active 2031-08-13 US11673970B2 (en) 2010-05-21 2020-10-23 Bi-specific fusion proteins
US17/212,270 Active 2032-02-19 US11814443B2 (en) 2010-05-21 2021-03-25 Bi-specific fusion proteins
US18/311,306 Pending US20230322951A1 (en) 2010-05-21 2023-05-03 Bi-Specific Fusion Proteins
US18/483,308 Pending US20240101713A1 (en) 2010-05-21 2023-10-09 Bi-Specific Fusion Proteins

Family Applications Before (5)

Application Number Title Priority Date Filing Date
US13/068,808 Active US9238080B2 (en) 2010-05-21 2011-05-20 Bi-specific fusion proteins
US13/112,907 Active 2031-11-01 US8691771B2 (en) 2010-05-21 2011-05-20 Bi-specific fusion proteins for tissue repair
US14/187,728 Active 2031-10-04 US9982060B2 (en) 2010-05-21 2014-02-24 Bi-specific fusion proteins
US14/967,980 Active US9718892B2 (en) 2010-05-21 2015-12-14 Method of treating myocardial infarction by administering a bi-specific fusion protein
US15/618,478 Active US10407512B2 (en) 2010-05-21 2017-06-09 Bi-specific fusion proteins

Family Applications After (5)

Application Number Title Priority Date Filing Date
US16/524,451 Active US10988547B2 (en) 2010-05-21 2019-07-29 Bi-specific fusion proteins
US17/078,978 Active 2031-08-13 US11673970B2 (en) 2010-05-21 2020-10-23 Bi-specific fusion proteins
US17/212,270 Active 2032-02-19 US11814443B2 (en) 2010-05-21 2021-03-25 Bi-specific fusion proteins
US18/311,306 Pending US20230322951A1 (en) 2010-05-21 2023-05-03 Bi-Specific Fusion Proteins
US18/483,308 Pending US20240101713A1 (en) 2010-05-21 2023-10-09 Bi-Specific Fusion Proteins

Country Status (9)

Country Link
US (11) US9238080B2 (en)
EP (1) EP2571992B1 (en)
JP (4) JP6200806B2 (en)
CN (1) CN103124788B (en)
AU (1) AU2011255238B2 (en)
BR (1) BR112012029611A2 (en)
CA (1) CA2800173C (en)
ES (1) ES2674567T3 (en)
WO (1) WO2011146902A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210040233A1 (en) * 2010-05-21 2021-02-11 Silver Creek Pharmaceuticals, Inc. Bi-Specific Fusion Proteins
US11879002B2 (en) 2015-10-02 2024-01-23 Silver Creek Pharmaceuticals, Inc. Bi-specific therapeutic proteins, in vivo methods of use thereof and encoding nucleic acids thereof

Families Citing this family (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102282168A (en) * 2008-11-18 2011-12-14 梅里麦克制药股份有限公司 Human serum albumin linkers and conjugates thereof
DK2396347T3 (en) 2009-02-11 2017-07-24 Albumedix As ALBUMIN VARIANTS AND CONJUGATES
BR112012009450A2 (en) 2009-10-30 2017-05-23 Novozymes Biopharma Dk As albumin variants
EP2536756B1 (en) * 2010-02-16 2018-04-25 MedImmune, LLC Hsa-related compositions and methods of use
CN102939304B (en) 2010-04-09 2017-04-19 阿尔布麦狄克斯公司 albumin derivatives and variants
US9045564B2 (en) 2011-02-15 2015-06-02 Medimmune, Llc HSA-related compositions and methods of use
CA2834516A1 (en) * 2011-04-28 2012-11-01 The Feinstein Institute For Medical Research Mfg-e8 and uses thereof
EP2705051A1 (en) 2011-05-05 2014-03-12 Novozymes Biopharma DK A/S Albumin variants
EP2780364A2 (en) 2011-11-18 2014-09-24 Eleven Biotherapeutics, Inc. Proteins with improved half-life and other properties
KR20140136934A (en) 2012-03-16 2014-12-01 노보자임스 바이오파마 디케이 에이/에스 Albumin variants
KR20220051197A (en) 2012-05-17 2022-04-26 익스텐드 바이오사이언시즈, 인크. Carriers for improved drug delivery
US9790264B2 (en) 2012-06-25 2017-10-17 The Brigham And Women's Hospital, Inc. Compounds and methods for modulating pharmacokinetics
CN104619727B (en) 2012-06-25 2019-04-05 布里格姆及妇女医院股份有限公司 Targeted therapy
WO2014036520A1 (en) 2012-08-30 2014-03-06 Merrimack Pharmaceuticals, Inc. Combination therapies comprising anti-erbb3 agents
KR20150082422A (en) 2012-11-08 2015-07-15 노보자임스 바이오파마 디케이 에이/에스 Albumin variants
WO2014139468A1 (en) * 2013-03-15 2014-09-18 Admark Healthcare, Llc Fusion protein molecules and method of use
ES2962694T3 (en) 2014-05-02 2024-03-20 Momenta Pharmaceuticals Inc Compositions and procedures related to manipulated Fc constructs
WO2015198199A1 (en) * 2014-06-23 2015-12-30 Novartis Ag Hsa-gdf-15 fusion polypeptide and use thereof.
US10588980B2 (en) 2014-06-23 2020-03-17 Novartis Ag Fatty acids and their use in conjugation to biomolecules
CN105273087A (en) * 2014-07-14 2016-01-27 复旦大学 NGF-Fc fusion protein and preparation method thereof
EP3220961B1 (en) 2014-10-22 2023-07-05 Extend Biosciences, Inc. Therapeutic vitamin d conjugates
US9789197B2 (en) 2014-10-22 2017-10-17 Extend Biosciences, Inc. RNAi vitamin D conjugates
US9616109B2 (en) 2014-10-22 2017-04-11 Extend Biosciences, Inc. Insulin vitamin D conjugates
WO2016168730A1 (en) 2015-04-17 2016-10-20 Merrimack Pharmaceuticals, Inc. Combination treatments with seribantumab
CN104946656B (en) * 2015-06-08 2018-08-17 吉林省农业科学院 A kind of people source basic fibroblast growth factor, tobacco chloroplast expression vector and production method
TWI744242B (en) 2015-07-31 2021-11-01 德商安美基研究(慕尼黑)公司 Antibody constructs for egfrviii and cd3
TWI829617B (en) 2015-07-31 2024-01-21 德商安美基研究(慕尼黑)公司 Antibody constructs for flt3 and cd3
TWI796283B (en) 2015-07-31 2023-03-21 德商安美基研究(慕尼黑)公司 Antibody constructs for msln and cd3
CA2989966A1 (en) 2015-08-20 2017-02-23 Albumedix A/S Albumin variants and conjugates
GB2542391A (en) * 2015-09-17 2017-03-22 Annexin Pharmaceuticals Ab Process of manufacture
US20190002516A1 (en) * 2015-12-21 2019-01-03 Hefei Lifeon Pharmaceutical Co., Ltd. Drug Design Method, Obtained Drug and Application Thereof
US20190365656A1 (en) * 2016-01-04 2019-12-05 Cour Pharmaceuticals Development Company, Inc. Particles encapsulating fusion proteins containing linked epitopes
RS62841B1 (en) 2016-02-03 2022-02-28 Amgen Res Munich Gmbh Bcma and cd3 bispecific t cell engaging antibody constructs
EA039859B1 (en) * 2016-02-03 2022-03-21 Эмджен Рисерч (Мюник) Гмбх Bispecific antibody constructs binding egfrviii and cd3
KR20180119570A (en) 2016-03-15 2018-11-02 메리맥 파마슈티컬즈, 인크. Treatment of ER +, HER2-, HRG + Breast Cancer with Combination Therapy Containing Anti-ErbB3 Antibody
IL263213B1 (en) * 2016-05-23 2024-01-01 Momenta Pharmaceuticals Inc Compositions and methods related to engineered fc constructs
US11220531B2 (en) 2017-01-06 2022-01-11 Janssen Biotech, Inc. Engineered Fc constructs
CA3068841A1 (en) 2017-07-03 2019-01-10 Torque Therapeutics, Inc. Fusion molecules targeting immune regulatory cells and uses thereof
AU2018307752A1 (en) * 2017-07-24 2020-03-12 Rutgers, The State University Of New Jersey Phosphatidylserine targeting fusion molecules and methods for their use
CN107880133A (en) * 2017-11-04 2018-04-06 海南大学 Corticotropin(ACTH) and type-1 insulin like growth factor fusion protein and preparation method thereof

Citations (65)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992008495A1 (en) 1990-11-09 1992-05-29 Abbott Biotech, Inc. Cytokine immunoconjugates
US5273966A (en) 1988-08-20 1993-12-28 Kabi Pharmacia Ab O-glycosylated IGF-1
WO1994028133A1 (en) * 1993-05-21 1994-12-08 Amgen Inc. Recombinant neu differentiation factors
WO1996033698A1 (en) 1995-04-24 1996-10-31 Northeastern University Compositions and methods useful for inhibiting cell death and for delivering an agent into a cell
US5632986A (en) 1991-05-09 1997-05-27 The University Of Washington Phospholipid-targeted thrombolytic agents
US5679771A (en) 1990-02-13 1997-10-21 Gropep Pty. Ltd. Method for treating intestinal diseases
EP0854884A1 (en) 1996-04-17 1998-07-29 Neurocrine Biosciences, Inc. Ligand inhibitors of insulin-like growth factor binding proteins and methods of use therefor
CA2286264A1 (en) 1997-04-04 1998-10-15 Genentech, Inc. Insulin-like growth factor agonist peptides
WO2000002587A1 (en) 1998-07-13 2000-01-20 Board Of Regents, The University Of Texas System Cancer treatment methods using therapeutic conjugates that bind to aminophospholipids
EP1141015A1 (en) 1999-01-06 2001-10-10 Genentech, Inc. Insulin-like growth factor (igf) i mutant variants
WO2002017951A1 (en) 2000-08-29 2002-03-07 Colorado State University Research Foundation Method for treating the central nervous system by administration of igf structural analogs
US6387663B1 (en) * 1998-07-31 2002-05-14 University Of Southern California Targeting pharmaceutical agents to injured tissues
US6566098B1 (en) 1990-09-14 2003-05-20 The United States Of America As Represented By The Department Of Health And Human Services DNA encoding truncated hepatocyte growth factor variants
EP1436316A2 (en) 2001-04-30 2004-07-14 Symbiontics, Inc. Subcellular targeting of therapeutic proteins
US20040213738A1 (en) 2003-03-31 2004-10-28 Susan Croll-Kalish CIRL3-Like proteins, nucleic acids, and methods of modulating CIRL3-L-mediated activity
US20050043236A1 (en) 2003-06-30 2005-02-24 Daly Thomas J. VEGF traps and therapeutic uses thereof
WO2005117973A2 (en) 2004-05-05 2005-12-15 Merrimack Pharmaceuticals, Inc. Bispecific binding agents for modulating biological activity
US20050287151A1 (en) 2003-09-30 2005-12-29 Glass David J Secreted protein therapeutics and uses thereof
WO2006004910A2 (en) 2004-06-28 2006-01-12 Transtarget Inc. Improved bispecific antibodies
WO2006003488A2 (en) 2004-07-07 2006-01-12 I.P. Randwijck B.V. Annexins, derivatives thereof, and annexin-cys variants, as well as therapeutic and diagnostic uses thereof
WO2006076525A2 (en) 2005-01-14 2006-07-20 Cytogen Corporation Combination cancer therapy with anti-psma antibodies
WO2006079120A2 (en) 2005-01-24 2006-07-27 Board Of Regents, The University Of Texas System Fc-fusion constructs binding to phosphatidylserine and their therapeutic use
WO2006091209A2 (en) 2005-02-23 2006-08-31 Merrimack Pharmaceuticals, Inc. Bispecific binding agents for modulating biological activity
US20060223753A1 (en) 2005-04-05 2006-10-05 Glass David J IGF-I and IGF-2 chimeric polypeptides and therapeutic uses thereof
WO2006128125A2 (en) 2005-05-27 2006-11-30 Five Prime Therapeutics, Inc. Methods of and compositions for stimulation of glucose uptake into muscle cells and treatment of diseases
US20060275254A1 (en) 2003-11-13 2006-12-07 Hanmi Pharm. Ind. Co., Ltd. Pharmaceutical composition comprising an immunoglobulin fc region as a carrier
WO2007021494A2 (en) 2005-08-12 2007-02-22 Human Genome Sciences, Inc. Albumin fusion proteins
WO2007044887A2 (en) 2005-10-11 2007-04-19 Transtarget, Inc. Method for producing a population of homogenous tetravalent bispecific antibodies
US20070110733A1 (en) 2002-04-23 2007-05-17 Lum Lawrence G Compositions and methods for stem cell delivery
US7226907B1 (en) 1998-12-21 2007-06-05 Zensun (Shanghai) Science & Technology Limited Cardiac muscle function and manipulation
US20070172811A1 (en) 2006-01-20 2007-07-26 Board Of Regents, The University Of Texas System Preserving hypoxic tissue
US20070224119A1 (en) 2004-10-21 2007-09-27 Igf Oncology Toxins and radionuclides coupled to IGF-1 receptor ligands for treatment of cancer
US20080050370A1 (en) 2006-03-17 2008-02-28 Scott Glaser Stabilized polypeptide compositions
AU2008200706A1 (en) 2001-10-16 2008-03-06 Biomarin Pharmaceutical Inc. Methods and compositions for targeting underglycosylated proteins across the blood brain barrier
US20080069823A1 (en) * 2001-02-21 2008-03-20 Alavita Pharmaceuticals, Inc. Attenuation of Reperfusion Injury
US20080095819A1 (en) 2004-12-21 2008-04-24 Robert Gourdie Composition and Methods for Promoting Wound Healing and tissue Regeneration
WO2008063424A2 (en) 2006-11-13 2008-05-29 The Brigham And Women's Hospital, Inc. Methods of promoting cardiac repair using growth factors fused to heparin binding sequences
US7396818B2 (en) 2003-05-19 2008-07-08 Seikagaku Corporation Sulfotransferase inhibitor
WO2008089567A1 (en) 2007-01-23 2008-07-31 F. Hoffmann-La Roche Ag Cancerous disease modifying antibodies
WO2008091209A1 (en) 2007-01-23 2008-07-31 Teknikbolaget K. Samuelsson Ab Nozzle means for extinguisher system
WO2008096158A2 (en) 2007-02-08 2008-08-14 Domantis Limited Antibody single variable domains against serum albumin
US20080241118A1 (en) 2001-10-16 2008-10-02 Zystor Therapeutics, Inc. Methods and Compositions for Targeting Proteins Across the Blood Brain Barrier
US7459541B2 (en) 1999-07-21 2008-12-02 University Of Southern California Matrix-targeted fusion polypeptides for tissue regeneration and wound healing
WO2008151005A2 (en) 2007-05-31 2008-12-11 Transtarget, Inc. Compositions and methods for tissue repair
WO2008155134A1 (en) 2007-06-21 2008-12-24 Technische Universität München Biological active proteins having increased in vivo and/or vitro stability
WO2009030720A2 (en) 2007-09-06 2009-03-12 Novozymes Biopharma Dk A/S Process for producing a recombinant protein
US7531318B2 (en) 2004-08-20 2009-05-12 Board Of Regents, The University Of Texas System Screening of agents for activity against ischemic myocardial insults
US20090214507A1 (en) 2004-08-20 2009-08-27 Regenerx Biopharmaceuticals, Inc. Method of treating preventing, inhibiting or reducing damage to cardiac tissue
WO2009126920A2 (en) 2008-04-11 2009-10-15 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
WO2010059315A1 (en) 2008-11-18 2010-05-27 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
US7837999B2 (en) 2005-01-07 2010-11-23 Regeneron Pharmaceuticals, Inc. Method of increasing muscle mass with IGF-1 fusion polypeptides
WO2011011071A2 (en) 2009-07-22 2011-01-27 Ipsen Pharma S.A.S. Analogues of insulin-like growth factor-1 (igf-1)
CA2768621A1 (en) 2009-07-22 2011-01-27 Ipsen Pharma S.A.S. Analogues of insulin-like growth factor-1 (igf-1) having amino acid substitution at position 59
US20110045007A1 (en) 2007-05-31 2011-02-24 Genmab A/S Fusion or linked proteins with extended half life
US20110274658A1 (en) 2007-04-05 2011-11-10 President And Fellows Of Harvard College Chimeric activators: quantitatively designed protein therapeutics and uses thereof
US8067357B2 (en) 2005-12-12 2011-11-29 Mosamedix B.V. Annexin derivatives suitable for pretargeting in therapy and diagnosis
US20110293579A1 (en) 2010-05-21 2011-12-01 Merrimack Pharmaceuticals, Inc. Bi-specific fusion proteins
WO2012078153A1 (en) 2010-12-08 2012-06-14 Viacyte, Inc. Agents and methods for inhibiting human pluripotent stem cell growth
US20120244163A1 (en) 2009-10-14 2012-09-27 Dyax Corp. Bispecific binding agents targeting igf-1r and erbb3 signalling and uses thereof
WO2013075066A2 (en) 2011-11-18 2013-05-23 Eleven Biotherapeutics, Inc. Proteins with improved half-life and other properties
WO2013086785A1 (en) 2011-12-15 2013-06-20 Qin Shulin Compound and composition having hypoglycemic effect and use thereof
US8748380B2 (en) 2009-10-30 2014-06-10 Novozymes Biopharma Dk A/S Albumin variants
CA2902744A1 (en) 2013-03-29 2014-10-02 Merrimack Pharmaceuticals, Inc. Cartilage-binding fusion proteins
EP2900255A2 (en) 2012-09-26 2015-08-05 Indiana University Research and Technology Corporation Insulin analog dimers
AU2015204540A1 (en) 2014-01-12 2016-08-04 Igf Oncology, Llc Fusion proteins containing insulin-like growth factor-1 and epidermal growth factor and variants thereof and uses thereof

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2190752A1 (en) 1994-05-24 1995-11-30 George N. Cox Modified insulin-like growth factors
GB2360771A (en) * 2000-03-28 2001-10-03 Antisoma Res Ltd Compounds for targeting
ATE464322T1 (en) * 2001-01-05 2010-04-15 Pfizer ANTIBODIES AGAINST THE INSULIN-LIKE GROWTH FACTOR RECEPTOR I
US7049286B2 (en) * 2001-08-30 2006-05-23 Diatos, S.A. Insulin conjugates and methods of use thereof
US7378393B2 (en) 2002-06-06 2008-05-27 Tze Chein Wun Recombinant anticoagulant proteins
WO2005066348A2 (en) * 2004-01-05 2005-07-21 Emd Lexigen Research Center Corp. Interleukin-12 targeted to oncofoetal fibronectin
US7855279B2 (en) 2005-09-27 2010-12-21 Amunix Operating, Inc. Unstructured recombinant polymers and uses thereof
JP2009525986A (en) * 2006-02-03 2009-07-16 メディミューン,エルエルシー Protein preparation
BRPI0707824A2 (en) 2006-02-15 2011-05-10 Imclone Systems Inc antigen-binding protein, and methods of neutralizing tyrosine kinase receptor activation, inhibiting angiogenesis, reducing tumor growth and producing an antigen-binding protein
US20100197890A1 (en) 2009-01-31 2010-08-05 Mctavish Hugh Anti-cancer protein-platinum conjugates
EP3857888A4 (en) 2018-10-06 2022-08-03 HFI Innovation Inc. Method and apparatus of shared merge candidate list region for video coding

Patent Citations (82)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5273966A (en) 1988-08-20 1993-12-28 Kabi Pharmacia Ab O-glycosylated IGF-1
US5679771A (en) 1990-02-13 1997-10-21 Gropep Pty. Ltd. Method for treating intestinal diseases
US6566098B1 (en) 1990-09-14 2003-05-20 The United States Of America As Represented By The Department Of Health And Human Services DNA encoding truncated hepatocyte growth factor variants
WO1992008495A1 (en) 1990-11-09 1992-05-29 Abbott Biotech, Inc. Cytokine immunoconjugates
US5632986A (en) 1991-05-09 1997-05-27 The University Of Washington Phospholipid-targeted thrombolytic agents
WO1994028133A1 (en) * 1993-05-21 1994-12-08 Amgen Inc. Recombinant neu differentiation factors
WO1996033698A1 (en) 1995-04-24 1996-10-31 Northeastern University Compositions and methods useful for inhibiting cell death and for delivering an agent into a cell
EP0854884A1 (en) 1996-04-17 1998-07-29 Neurocrine Biosciences, Inc. Ligand inhibitors of insulin-like growth factor binding proteins and methods of use therefor
CA2286264A1 (en) 1997-04-04 1998-10-15 Genentech, Inc. Insulin-like growth factor agonist peptides
WO2000002587A1 (en) 1998-07-13 2000-01-20 Board Of Regents, The University Of Texas System Cancer treatment methods using therapeutic conjugates that bind to aminophospholipids
US6387663B1 (en) * 1998-07-31 2002-05-14 University Of Southern California Targeting pharmaceutical agents to injured tissues
US7226907B1 (en) 1998-12-21 2007-06-05 Zensun (Shanghai) Science & Technology Limited Cardiac muscle function and manipulation
US7612164B2 (en) 1998-12-21 2009-11-03 Zensun (Shanghai) Science and Technologies Ltd. Cardiac muscle function and manipulation
EP1141015A1 (en) 1999-01-06 2001-10-10 Genentech, Inc. Insulin-like growth factor (igf) i mutant variants
US20090093407A1 (en) 1999-07-21 2009-04-09 Hall Frederick L Matrix-targeted fusion polypeptides for tissue regeneration and wound healing
US7459541B2 (en) 1999-07-21 2008-12-02 University Of Southern California Matrix-targeted fusion polypeptides for tissue regeneration and wound healing
WO2002017951A1 (en) 2000-08-29 2002-03-07 Colorado State University Research Foundation Method for treating the central nervous system by administration of igf structural analogs
US20080069823A1 (en) * 2001-02-21 2008-03-20 Alavita Pharmaceuticals, Inc. Attenuation of Reperfusion Injury
EP1436316A2 (en) 2001-04-30 2004-07-14 Symbiontics, Inc. Subcellular targeting of therapeutic proteins
US20080241118A1 (en) 2001-10-16 2008-10-02 Zystor Therapeutics, Inc. Methods and Compositions for Targeting Proteins Across the Blood Brain Barrier
AU2008200706A1 (en) 2001-10-16 2008-03-06 Biomarin Pharmaceutical Inc. Methods and compositions for targeting underglycosylated proteins across the blood brain barrier
US7576186B2 (en) 2002-04-23 2009-08-18 Roger Williams Hospital Compositions and methods for stem cell delivery
US20100055115A1 (en) 2002-04-23 2010-03-04 Lum Lawrence G Compositions and methods for stem cell delivery
US20070110733A1 (en) 2002-04-23 2007-05-17 Lum Lawrence G Compositions and methods for stem cell delivery
US20040213738A1 (en) 2003-03-31 2004-10-28 Susan Croll-Kalish CIRL3-Like proteins, nucleic acids, and methods of modulating CIRL3-L-mediated activity
US7396818B2 (en) 2003-05-19 2008-07-08 Seikagaku Corporation Sulfotransferase inhibitor
US20050043236A1 (en) 2003-06-30 2005-02-24 Daly Thomas J. VEGF traps and therapeutic uses thereof
US20050287151A1 (en) 2003-09-30 2005-12-29 Glass David J Secreted protein therapeutics and uses thereof
US20060275254A1 (en) 2003-11-13 2006-12-07 Hanmi Pharm. Ind. Co., Ltd. Pharmaceutical composition comprising an immunoglobulin fc region as a carrier
WO2005117973A2 (en) 2004-05-05 2005-12-15 Merrimack Pharmaceuticals, Inc. Bispecific binding agents for modulating biological activity
US20060018897A1 (en) 2004-06-28 2006-01-26 Transtarget Inc. Bispecific antibodies
US20090068181A1 (en) 2004-06-28 2009-03-12 Transtarget, Inc. Bispecific antibodies
WO2006004910A2 (en) 2004-06-28 2006-01-12 Transtarget Inc. Improved bispecific antibodies
WO2006003488A2 (en) 2004-07-07 2006-01-12 I.P. Randwijck B.V. Annexins, derivatives thereof, and annexin-cys variants, as well as therapeutic and diagnostic uses thereof
US20090214507A1 (en) 2004-08-20 2009-08-27 Regenerx Biopharmaceuticals, Inc. Method of treating preventing, inhibiting or reducing damage to cardiac tissue
US7531318B2 (en) 2004-08-20 2009-05-12 Board Of Regents, The University Of Texas System Screening of agents for activity against ischemic myocardial insults
US20070224119A1 (en) 2004-10-21 2007-09-27 Igf Oncology Toxins and radionuclides coupled to IGF-1 receptor ligands for treatment of cancer
US20080095819A1 (en) 2004-12-21 2008-04-24 Robert Gourdie Composition and Methods for Promoting Wound Healing and tissue Regeneration
US8445434B2 (en) 2005-01-07 2013-05-21 Regeneron Pharmaceuticals, Inc. IGF-1 fusion polypeptides and therapeutic uses thereof
US8158581B2 (en) 2005-01-07 2012-04-17 Regeneron Pharmaceuticals, Inc. IGF-1 fusion polypeptides and therapeutic uses thereof
US7837999B2 (en) 2005-01-07 2010-11-23 Regeneron Pharmaceuticals, Inc. Method of increasing muscle mass with IGF-1 fusion polypeptides
WO2006076525A2 (en) 2005-01-14 2006-07-20 Cytogen Corporation Combination cancer therapy with anti-psma antibodies
WO2006079120A2 (en) 2005-01-24 2006-07-27 Board Of Regents, The University Of Texas System Fc-fusion constructs binding to phosphatidylserine and their therapeutic use
WO2006091209A2 (en) 2005-02-23 2006-08-31 Merrimack Pharmaceuticals, Inc. Bispecific binding agents for modulating biological activity
US20060223753A1 (en) 2005-04-05 2006-10-05 Glass David J IGF-I and IGF-2 chimeric polypeptides and therapeutic uses thereof
WO2006128125A2 (en) 2005-05-27 2006-11-30 Five Prime Therapeutics, Inc. Methods of and compositions for stimulation of glucose uptake into muscle cells and treatment of diseases
US20070054851A1 (en) * 2005-05-27 2007-03-08 Junyu Lin Methods of and compositions for stimulation of glucose uptake into muscle cells and treatment of diseases
WO2007021494A2 (en) 2005-08-12 2007-02-22 Human Genome Sciences, Inc. Albumin fusion proteins
WO2007044887A2 (en) 2005-10-11 2007-04-19 Transtarget, Inc. Method for producing a population of homogenous tetravalent bispecific antibodies
US8067357B2 (en) 2005-12-12 2011-11-29 Mosamedix B.V. Annexin derivatives suitable for pretargeting in therapy and diagnosis
US20070172811A1 (en) 2006-01-20 2007-07-26 Board Of Regents, The University Of Texas System Preserving hypoxic tissue
US20080050370A1 (en) 2006-03-17 2008-02-28 Scott Glaser Stabilized polypeptide compositions
WO2008063424A2 (en) 2006-11-13 2008-05-29 The Brigham And Women's Hospital, Inc. Methods of promoting cardiac repair using growth factors fused to heparin binding sequences
WO2008089567A1 (en) 2007-01-23 2008-07-31 F. Hoffmann-La Roche Ag Cancerous disease modifying antibodies
WO2008091209A1 (en) 2007-01-23 2008-07-31 Teknikbolaget K. Samuelsson Ab Nozzle means for extinguisher system
WO2008096158A2 (en) 2007-02-08 2008-08-14 Domantis Limited Antibody single variable domains against serum albumin
US20110274658A1 (en) 2007-04-05 2011-11-10 President And Fellows Of Harvard College Chimeric activators: quantitatively designed protein therapeutics and uses thereof
US20100291080A1 (en) 2007-05-31 2010-11-18 Lee Randall J Compositions and methods for tissue repair
US20110045007A1 (en) 2007-05-31 2011-02-24 Genmab A/S Fusion or linked proteins with extended half life
WO2008151005A2 (en) 2007-05-31 2008-12-11 Transtarget, Inc. Compositions and methods for tissue repair
WO2008155134A1 (en) 2007-06-21 2008-12-24 Technische Universität München Biological active proteins having increased in vivo and/or vitro stability
WO2009030720A2 (en) 2007-09-06 2009-03-12 Novozymes Biopharma Dk A/S Process for producing a recombinant protein
WO2009126920A2 (en) 2008-04-11 2009-10-15 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
US20110059076A1 (en) 2008-11-18 2011-03-10 Mcdonagh Charlotte Human serum albumin linkers and conjugates thereof
WO2010059315A1 (en) 2008-11-18 2010-05-27 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
WO2011011071A2 (en) 2009-07-22 2011-01-27 Ipsen Pharma S.A.S. Analogues of insulin-like growth factor-1 (igf-1)
CA2768621A1 (en) 2009-07-22 2011-01-27 Ipsen Pharma S.A.S. Analogues of insulin-like growth factor-1 (igf-1) having amino acid substitution at position 59
US20120244163A1 (en) 2009-10-14 2012-09-27 Dyax Corp. Bispecific binding agents targeting igf-1r and erbb3 signalling and uses thereof
US8748380B2 (en) 2009-10-30 2014-06-10 Novozymes Biopharma Dk A/S Albumin variants
US9718892B2 (en) * 2010-05-21 2017-08-01 Merrimack Pharmaceuticals, Inc. Method of treating myocardial infarction by administering a bi-specific fusion protein
US20110293579A1 (en) 2010-05-21 2011-12-01 Merrimack Pharmaceuticals, Inc. Bi-specific fusion proteins
US8691771B2 (en) * 2010-05-21 2014-04-08 Merrimack Pharmaceuticals, Inc. Bi-specific fusion proteins for tissue repair
US10407512B2 (en) * 2010-05-21 2019-09-10 Silver Creek Pharmaceuticals, Inc. Bi-specific fusion proteins
US9982060B2 (en) * 2010-05-21 2018-05-29 Merrimack Pharmaceuticals, Inc. Bi-specific fusion proteins
US9238080B2 (en) * 2010-05-21 2016-01-19 Merrimack Pharmaceuticals, Inc. Bi-specific fusion proteins
WO2012078153A1 (en) 2010-12-08 2012-06-14 Viacyte, Inc. Agents and methods for inhibiting human pluripotent stem cell growth
WO2013075066A2 (en) 2011-11-18 2013-05-23 Eleven Biotherapeutics, Inc. Proteins with improved half-life and other properties
US20140315817A1 (en) 2011-11-18 2014-10-23 Eleven Biotherapeutics, Inc. Variant serum albumin with improved half-life and other properties
WO2013086785A1 (en) 2011-12-15 2013-06-20 Qin Shulin Compound and composition having hypoglycemic effect and use thereof
EP2900255A2 (en) 2012-09-26 2015-08-05 Indiana University Research and Technology Corporation Insulin analog dimers
CA2902744A1 (en) 2013-03-29 2014-10-02 Merrimack Pharmaceuticals, Inc. Cartilage-binding fusion proteins
AU2015204540A1 (en) 2014-01-12 2016-08-04 Igf Oncology, Llc Fusion proteins containing insulin-like growth factor-1 and epidermal growth factor and variants thereof and uses thereof

Non-Patent Citations (118)

* Cited by examiner, † Cited by third party
Title
Adderson, E., et al., "Molecular analysis of polyreactive monoclonal antibodies from rheumatic carditis: human anti-N-accytlglucosamine/anti-myosin antibodyV region genes", J Immunol, 161(4):2020-2031, (Aug. 15, 1998).
Andrades, et al., "Engineering, expression, and renaturation of a collagen-targeted human bFGF fusion protein," Growth Factors, 18:261-275, (Aug. 1999).
Askari, et al., "Effect of stromal-cell-derived factor 1 on stem-cell homing and tissue regeneration in ischaemic cardiomyopathy," Mechanisms of Disease, 362: 697-703, (Aug. 30, 2003).
Bai et al., "Tracking Long-Term Survival of Intramyocardially Delivered Human Adipose Tissue-Derived Stem Cells Using Bioluminescence Imaging", Moleculr Imaging and Biology, 13 pages (Aug. 21, 20120).
Barbas, S., et al., "Human autoantibody recognition of DNA," Proc Natl Acad Sci US A, 92(7):2529-2533, (Mar. 28, 1995).
Bauwens, C., e al., "Geometric Control of Cardiomyogenic Induction in Human Pluripotent Stem Cells", Tissue Eng., Part A, (Apr. 25, 2011).
Bersell, et al., "Neuregulinl/ErbB4 signaling induces cardiomyocyte proliferation and repair of heart injury," Cell, 138:257-270, (Jul. 24, 2009).
Black, S., "In vivo models of myocardial ischemia and reperfusion injury: application to drug discovery and evaluation," J. Pharmacol. Toxicol. Methods 43(2):153-167, (Mar.-Apr. 2000).
Bock-Marquette, et al., "Thymosin B4 activates integrin-linked kinase and promotes cardiac cell migration, survival and cardiac repair," Nature, 432:466-472, (Nov. 25, 2004).
Buerke, et al., "Cardioprotective effect of insulin-like growth factor I in myocardial ischemia followed by reperfusion," Proc. Natl. Acad. Sci. USA, 92: 8031-8035, (Aug. 1995).
Bujak Met al. "The role of TGF beta signaling in myocardial infection and cardiac remodeling" Cardiovasc. Res. (2007) May 1, 74(2),: pp. 184-195.
Burchfield, et al., "Interleukin-10 from transplanted bone marrow mononuclear cells contributes to cardiac protection after myocardial infarction," Circulation Research, 15 pages, (Mar. 23, 2011 ).
Burchfield, et al., "Role of paracrine factors in stem and progenitor cell mediated cardiac repair and tissue fibrosis," Fibrogenesis and Tissue Repair, 1( 4): 1-11, (2008).
Burchfield, et al., "The cytoprotective effects of tumor necrosis factor are conveyed through tumor necrosis factor receptor-associated factor 2 in the heart," Circulation Heart Failure, 16 pages, (Jan. 2010).
Chen et al., "Effects of Receptor Binding on Plasma Half-Life of Bifunctional Transferrin Fusion Proteins", Molecular Pharmaceutics 8: 457-65 (Feb. 1, 2011).
Chen, et al., "Localization of monoclonal antibody TNT-I in experimental kidney infarction of the mouse," FASEB J., 4 (12):3033-3039, (Sep. 1, 1990).
Chimenti, et al., "Myocardial infarction: animal models," Methods. Mol. Med., 98:217-226, (2004).
Christman, et al., "Enhanced neovasculature formation in ischemic myocardium following delivery of pleiotrophin plasmid in a biopolymer,"Biomaterials, 26: 1139-1144 (2005).
Davis, "Local myocardial insulin-like growth factor 1 (IGF-1) delivery with biotinylated peptide nanofibers improves cell therapy for myocardial infarction," Proc. Natl. Acad. Sci USA, 103(21):8155-8160, (May 23, 2006).
Davletov, A. & Sudhof, T., "A Single C2 Domain from Synaptotagmin I is Sufficient for High Affinity Ca2+/Phospholipid Binding", J. Biol. Chem., 268(35):26386-2690, (Dec. 15, 1993).
Dolden-Martelli et al., "A Mathematical model for the Rational Design of Chimeric Ligands in Selective Drug Therapies", CPT: Pharmacometrics & Systems Pharmacology (2013), 2, ep26, Feb. 13, 2013.
Dom II, M.D., "Periostin and myocardial repair, regeneration, and recovery," The New England Journal of Medicine, 357(15):1552-1554, (Oct. 11, 2007).
Dubaquie, Y. et al., "Total Alinine-Scanning Mutagenesis of Insulin-Like Growth Factor I (IGF-I) Identifies Differential Binding Epitopes for IGFBP-1 and IGFBP-3", Biochemistry, 1999, 38, 6386-6396.
Dumont, et al., "Cardiomyocyte Death Induced by Myocardial Ischemia and Reperfusion: Measurement With Recombinant Human Annexin-V in a Mouse Model ," Circulation 102(13):1564-1568, (Sep. 26, 2000).
Engel, et al., "FGF1/p38 MAP kinase inhibitor therapy induces cardiomyocyte mitosis, reduces scarring, and rescues function after myocardial infarction," PNAS, 103(42):15546-15551, (Oct. 17, 2006).
Epa, V.C. et al., "Model for the Complex Between the Insulin-Like Growth Factor 1 and Its Receptors: Towards Designing Antagonists for the IGF-1 Receptor", Protein Engineering, Design & Selection, vol. 19, No. 8, pp. 377-384, 2006.
George, et al., "Typhostin AG-556 reduces myocardial infarct size and improves cardiac performance in the rat," Experimental and Molecular Patholo{IV, 74:314-318 (2003).
Gnecchi, et al., "Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement," The FASEB Journal, 20:661-669, (Apr. 2006).
Gnecchi, et al., "Paracrine mechanisms in adult stem cell signaling and therapy," Adult Stem Cells and Paracrine Effects, 1204-1219, (Jan. 9, 2010).
Greenberg, et al., "Chapter 7. Mouse models of ischemic angiogenesis and ischemia-reperfusion injury," Methods Enzymol., 444: 159-174, (2008).
Gripenberg, et al., "A Solid Phase Enzyme-linked Immunosorbent Assay (ELISA) for the Demonstration of Antibodies against Denatured, Single-stranded DNA in Patient Sera," Scand. J. Immunol., 7(2):151-157, (Feb. 1978).
Han, et al., "Refolding of a recombinant collagen-targeted TGF-B2 fusion protein expressed in Escherichia coli," Protein Expression and Purification, 11: 169-178 (1997).
Hashino, K., et al., "A 31-kDa Recombinant Fibronectin Cell-Binding Domain Fragment: Its Binding to Receptor, Cell Adhesive Activity, and Fusion Proteins," J. Biochem., 119(4):604-609, (Apr. 4, 1996).
Hausenloy et al., "Cardioprotective Growth Factors", Cardiovascular Research, 83: 179-194. (Feb. 13, 2009).
Hefta, et al., "Measuring Affinity Using Biosensors", in "Antibody engineering: A Practical Approach", pp. 99-116, Dxford University Press, 1996, Edited by McCafferty et al., (Hames B.D.eds).
Henson E. S. et al., "Surving Cell Death Through Epidermal Growth Factor (EGF) Signal Transduction pathways: Implication for Cancer Therapy", Cellular Signaling (2006) 18; pp. 2089-2097.
Hinkel et al., "Thymosin B4 is an essential paracrine factor of embryonic endothelial progenitor cell-mediated cardioprotection," Circulation, 2232-2240 (Apr. 29, 2008).
Hoberg, E., et al., "Monoclonal antibodies specific for human cardiac myosin: selection, characterization and experimental myocardial infarct imaging," Eur Heart J., 9(3):328-236, (Mar. 1988).
Hofstra, et al., "Visualisation of cell death in vivo in patients with acute myocardial infarction," The Lancet, 356 (9225)209-212, (2000).
Hsieh et al., "Local controlled intramyocardial delivery of plateet-derived growth factor improves postinfarction ventricular function without pulmonary toxicity," Circulation, 637-644, (Aug. 15, 2006).
Hu et al., Stromal cell-derived factor-I a confers protection against myocardial ischemia/reperfusion injury, Molecular Cardiology, 654-663, (Aug. 7, 2007).
Ieda, et al., "Cardiac fibroblasts regulate myocardial proliferation through BI integrin signaling," Developmental Cell, 16: 233-244 (Feb. 17, 2009).
Igarashi, K., et al., "Specific binding of a synthetic peptide derived from an antibody complementarity determining region to phosphatidylserine," J Biochem.,117(2):452-457, (Feb. 1995).
Ishikawa, et al., "Production of biologically active epidermal growth factor fusion protein with high collagen affinity," J. Biochem., 129(4): 627-633 (2001).
Jeon, et al., "Long-term and zero-order release of basic fibroblast growth factor from heparin-conjugated poly(L-lactide-co-glycolide) nanospheres and fibrin gel," Biomaterials, 27:1598-1607 (2006).
Kanashiro-Takeuchi, et al., "Cardioprotective effects of growth hormone-releasing hormone agonist after myocardial infarction," PNAS, 107(6):2604-2609, (Feb. 9, 2010).
Kardami, et al., "Fibroblast growth factor-2 and cardioprotection," Heart Fail Rev., 12:267-277 (2007).
Kawase Y. et al. "Construction and characterization of a fusion protein with epidermal growth factor and the cell-binding domain of fibronectin" FEBS letters, 298(2-3),: 126-128, 1992.
Kenis et al., Annexin A5: shifting from a diagnostic towards a therapeutic realm, Cell. Mol. Life Sci., vol. 64:2859-2862 (2007) (Year: 2007). *
Kenis, H., et al., "Annexin A5 uptake in ischemic myocardium: demonstration of reversible phosphatidylserine externalization and feasibility of radionuclide imaging," J Nucl Med., 51(2):259-67, (Feb. 2010).
Kenis, H., et al., "Cell Surface-Expressed Phosphatidylserine and Annexin A5 Open a Novel Portal of Cell Entry", J Biol Chem., 279(50):52623-52629, Epub Sep. 20, 2004, (Dec. 10, 2004).
Khaw, B., et al., "Monoclonal antibody to cardiac myosin: imaging of experimental myocardial infarction," Hybridoma, 3(1 ): 11-23, (1984).
King et al., "Production and Characterization of Recombinant Insulin-Like Growth Factor-I (IGF-I) and Potent Analogues of IGF-I, with Gly or Arg Substituted for Glu3, Following their Expression in Escherichia colu as Fusion Proteins", Journal of Molecular Endocrinology, (1992) 8, pp. 29-41.
Klopsch et al., "Intracardiac Injection of Erythropoietin Induces Stem Cell Recruitment and Improves Cardiac Functions in a Rat Myocardial Infraction Model", J. Cell. Mol. Med. 13(4): 664-679, (Apr. 2009).
Ko, Y., et al., "Gene delivery into ischemic myocardium by double-targeted lipoplexes with anti-myosin antibody and TAT peptide," Gene Ther., 16(1):52-9. Epub Aug. 14, 2008, (Jan. 2009).
Kobayashi, et al., "Effect of atrial natriuretic peptide on ischemia-reperfusion injury in a porcine total hepatic vascular exclusion model," World J. Gastroenterol., 13(25):3487-3492, (Jul. 7, 2007).
Kuhn, et al., "Periostin induces proliferation of differentiated cardiomyocytes and promotes cardiac repair," Nature Medicine, 13(8):962-969, (Aug. 2007).
Kuramochi, "Cardiac Endothelial Cells Regulate Reactive Oxygen Species-induced Cardiomyocyte Apoptosis through Neuregulin-1_/erbB4 Signaling*," J. Biol. Chem., 279(49): 51141-51147, (2004).
Laajoki, L et al., "Solution Structure and Backbone Dynamics of Long-[ARG3] Insulin-Like Growth Factor-I", Journal of Biological Chemistry, vol. 275, No. 14, pp. 10009-10015, Apr. 7, 2000.
Laroche-Traineau, J., et al., "A human monoclonal antibody obtained from EBV-transformed B cells with specificity for myosin," Br J Haematol., 91 ( 4):951-962, (Dec. 1995).
Laroche-Traineau, J., et al., "Analysis of the V genes coding for a monospecific human antibody to myosin and functional expression of single chain Fv fragments," FEBS Lett., 460(1 ):86-92, (Oct. 22, 1999).
Laroche-Traineau, J., et al., "Three-step purification of bacterially expressed human single-chain Fv antibodies for clinical applications," J Chromatogr B Biomed Sci Appl., 737(1-2):107-117, (Jan. 14, 2000).
Liang, W., et al., "ATP-containing immunoliposomes specific for cardiac myosin," Curr Drug Deliv., 1(1):1-7, (Jan. 2004).
Liu, et al., "Neuregulin-1/erbB-activation improves cardiac function and survival in models of ischemic, dilated, and viral cardiomyopathy," Journal of the American College ofCardiolo2:v, 48(7):1438-1447, (Oct. 3, 2006).
Loddick, S. et al., "Displacement of Insulin-Like Growth Factors from their Binding Proteins as a Potental Treatment for Stroke", PNAS, vol. 95, pp. 1894-1898, Feb. 1998.
Lorts et al., "Genetic Manipulation of Periostin Expression in the Heart Does Not Affect Myocyte Content, Cell Cycle Activity, or Cardiac Repair", UltraRapid Communications, e1-e7, (Jan. 2, 2009).
Marshall, K.W. & Marks, J.D. "Engineering and characterization of a novel fusion protein incorporating B7.2 and an anti-ErbB-2 single-chain antibody fragment for the activation of Jurkat T cells," Journal ofImmunotherapy. Hagerstown, Md. : 1997) 24: 27-36 (2001).
Mihardja, et al., "Targeted in vivo extracellular matrix formation promotes neovascularization in a rodent model of myocardial infarction," PLoS One, 5(4):e10384 (8 pages), (Apr. 2010).
Mira, et al., "Inhibition of cytosolic phospholipase A2 by annexin V in differentiated permeabilized HL-60 cells. Evidence of crucial importance of domain I type II Ca2+-binding site in the mechanism of inhibition," J. Biol Chem., 272(16): 104 74-10482, (Apr. 18, 1997).
Miranda, et al., "Endothelium-dependent and -independent hepatic artery vasodilatation is not impaired in a canine model of liver ischemia-reperfusion injury," Braz. J. Med. Biol. Res., 40(6):857-865, (Jun. 2007).
Murray and Brown, "Measurement of association constants in ELISA. Reactions between solid-phase antibody and fluid-phase biotinylated antigen," J. Immunol. Methods., 127(1):25-28 (Feb. 20, 1990).
Nedelman, M., et al., "Rapid infarct imaging with a technetium-99m-labeled antimyosin recombinant single-chain Fv: evaluation in a canine model of acute myocardial infarction," J Nucl Med., 34(2):234-241, (Feb. 1993).
Nelson, P., et al. "Characterization of anti-myosin monoclonal antibodies," Hybridoma (Larchmt), 24(6):314-318, (Dec. 2005).
Nimni, "Polypeptide growth factors: targeted delivery systems," Biomaterials, 18(18):1201-1225, (1997).
Nishi, et al., "Collagen-Binding Growth Factors: Production and Characterization of functional fusion proteins having a collagen-binding domain," Proc. Natl. Acad. Sci. USA, 95:7018-7023, (Jun. 1998).
Novo Nordisk Pharmatech A/S "How Insulin and IGF-1 Bind to the Receptors" retrieved from http://novonordiskpharmatech.com/how-insulin0and-igf-1-bind-to-their-receptors/ ;retrieved Dec. 16, 2016.
O'Sullivan et al., "Potent Long-Term Cardioprotective Effects of Single Low-Dose Insulin-Like Growth Factor-1 Treatment Postmyocardial Infarction", American Heart Association-Circulation: Cardiovascular Interventions, 1:327-335, Jun. 28, 2011.
O'Sullivan et al., "Potent Long-Term Cardioprotective Effects of Single Low-Dose Insulin-Like Growth Factor-1 Treatment Postmyocardial Infarction", American Heart Association—Circulation: Cardiovascular Interventions, 1:327-335, Jun. 28, 2011.
Pak, K., et al., "An instant kit method for labeling antimyosin Fab' with technetium-99m: evaluation in an experimental myocardial infarct model," J. Nucl Med., 33(1):144-149, (Jan. 1992).
Peter, K., et al., "Construction and Functional Evaluation of a Single-Chain Antibody Fusion Protein with Fibrin Targeting and Thrombin Inhibition after Activation by Factor Xa", Circulation, 101(10):1158-1164, (Mar. 14, 2000).
Pietronave et al., "Agonist Monoclonal Antibodies Against HGF Receptor Protect Cardiac Muscle Cells from Apoptosis", Am J Physiol Heart circ Physiol, 298:H1155-H1165, (Jan. 8, 2010).
Prior, et al. "Cytotoxic Activity of a Recombinant Fusion Protein between Insulin-like Growth Factor I and Pseudomonas Exotoxin," Cancer, 174-180 (1991 ).
Rosenthal, et al., "Growth factor enhancement of cardiac regeneration," Cell Transplantation, 15(1):S41-S45, (2006).
Saxena, et al., "Stromal cell-derived factor-la is cardioprotective after mocardial infarction," Molecular Cardiology, 2224-2231, (2008).
Schutters et al., Phosphatidylserine targeting for diagnosis and treatment of human disease, Apoptosis, vol. 15:1072-1082 (May 4, 2010) (Year: 2010). *
Schutters, K. & Reutelingsperger, CPM. "Phosphatidylserine Targeting for Diagnosis and Treatment of Human Diseases", Apoptosis: An International Journal on Programmed Cell Death. 15:1072-82, (May 4, 2010).
Scott, et al. "Aiming for the heart: targeted delivery of drugs to diseased cardiac tissue," Expert Opinion on Drug Delivery, 5:459-70, (2008).
Scott, et al. "Targeting VEGF-encapsulated immunoliposomes to MI heart improves vascularity and cardiac function", The FASEB Journal: Research Communication, vol. 23, pp. 3361-3367, (Oct. 2009).
Scott, R.C. et al.,"Targeted Delivery of Antibody Conjugated Lipsomal Drug Carriers to Rat Myocardial Infarction", Biotechnology, 96:795-802, (Mar. 1, 2007).
Segers, et al., "Protein therapeutics for cardiac regeneration after myocradial infarction," J. of Cardiovasc. Trans. Res., 9 pages, (Jul. 7, 2010).
Shan, et al., "Overexpression of TRPC3 increases apoptosis but not necrosis in response to ischemia-reperfusion in adult mouse cardiomyocytes," Am. J. Physiol. Cell. Physiol., 294(3):833-841, (Mar. 2008).
Shin, et al. "Functional properties of antibody insulin-like growth factor fusion proteins," The Journal of Biological Chemistry, 269: 4979-8,5 (1994).
Shin, S.U. & Morrison, S.L., "Expression and characterization of an antibody binding specificity joined to insulin-like growth factor 1: potential applications for cellular targeting," Proceedings of the National Academy of Sciences of the United States of America, 87:5322-6, (1990).
Simeonova, P. et al., "Identification of Human Ventricular Myosin Heavy Chain Fragments with Monoclonal Antibody 2F4 in Human Sera after Myocardial Necrosis", Clin. Chim. Acta., 201(3):207-221, (Sep. 30, 1991).
Stamm, et al., Human ortholog to mouse gene imap38 encoding an ER-localizable G-protein belongs to a gene family clustered on chromosome 7q32-36, Gene vol. 282: 159-167 (2002).
Stokes, et al., "A simple, rapid ELISA method for the detection of DNA antibodies," J. Clin. Pathol., 35(5):566-573, (May 1982).
Suleiman, et al., "Apoptosis and the cardiac action of insulin-like growth factor I," Pharmacoloe:v and Therapeutics, 114:278-294, (2007).
Sutton, R., et al., "Structure of the first C2 domain of synaptotagmin I: a novel Ca2+/phospholipid-binding fold," Cell, 80(6):929-938, (Mar. 24, 1995).
Tomas, F.M. et al., "IGF-I Variants Which Bond Poorly to IGF-Binding Proteins Show More Potent and Prolonged Hypoglycemic Action that Native IGF-I in Pigs and Marmoset Monkeys", Journal of Endocrinology (1997), 155, 377-386.
Tuan et al., Engineering, expression and renaturation of targeted TGF-beta fusion proteins: Connect Tissue Res. 1996:34(1 ): 1-9.
Ueda, et al., "A potential cardioprotective role of hepatocyte growth factor in myocardial infarction in rats," Cardiovascular Research, 51:41-50, (2001).
Umeda, M., et al., "Effective production of monoclonal antibodies against phosphatidylserine: stereo-specific recognition of phosphatidylserine by monoclonal antibody," J Immunol., 143(7):2273-2279, (Oct. 1, 1989).
Ungethum, et al., "Engineered annexin A5 variants have impaired cell entry for molecular imaging of apoptosis using pretargeting strategies," J Biol Chem., 286(3):1903-10. Epub Nov. 15, 2010 (Jan. 21, 2011).
Urbanek Ket al., "Myocardial regeneration by activation of multipotent cardiac stem cells in ischemic heart failure," Proc. Natl. Acad. Sci. USA, 102(24):8692-8697, (Jun. 14, 2005).
Wang et al., "Degradable PLGA Scaffolds with Basic Fibroblast Growth Factor", Texas Heart Institute Journal, 36(2): 89-97 (Apr. 2009).
Wassaf, et al., "High-throughput affinity ranking of antibodies using surface plasmon resonance microarrays," Anal. Biochem., 351(2):241-253, (Apr. 15, 2006).
Winter et al., "A New Bioassay for the Immunocytokine L19-IL2 for Simultaneous Analysis of Both Functional Moieties", Journal of Pharmaceutical and Biomedical Analysis, 54:81-6 (Jan. 5, 2011).
Yang L. et al., "Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population", Nature. May 22, 2008; 453(7194):524-8. Epub Apr. 23, 2008.
Yeghiazarians et al., "Injection of Bone Marrow Cell Extract into Infarcted Hearts Results in Functional Improvement Comparable to Intact Cell Therapy", The American Society of Gene Therapy, 17(7):1250-1256, (Jul. 2009).
Zaruba, et al., "Synergy between CD26/DPP-IV inhibition and G-CSF improves cardiac function after acute myocardial infarction," Cell Stem Cell, 4:313-323, (Apr. 3, 2009).
Zbinden, et al., "Interanimal variability in preexisting collaterals is a major factor determining outcome in experimental angiogenesis trials," Am. J. Physiol. Heart Circ. Physiol., 292(4): H1891-H1897, (Apr. 2007).
Zentilin, et al., "Cardiomyocyte VEGFR-1 activation by VEGF-B induces compensatory hypertrophy and preserves cardiac function after myocardial infarction," The F ASEB Journal, 24:1467-1478, (May 2010).
Zhang et al., "Collagen-Targeting Vascular Endothelial Growth Factor Improves Cardiac Performance After Myocradial Infarction", Circulation, vol. 119(13):1776-84 (epub Mar. 23, 2009).
Zhang et al., Collagen-targeting vascular endothelial growth factor improves cardiac performance after myocardial infarction, Circulation, vol. 119(13):1776-84 (epub Mar. 23, 2009) (Year: 2009). *
Zhao et al., J. Nucl Med, vol. 47(8):1367-1374 (Aug. 2006).
Zhao, et al., "Neuregulins promote survival and growth of cardiac myocytes," The Journal of Biological Chemistry, 273(17):0261-10269, (Apr. 24, 1998).
Zhao, et al., "Recruitment of endogenous stem cells for tissue repair," Macromolecular Bioscience, 8:836-842, (2008).
Ziegler, M. et al., "The bispecific SDF 1-GPVI fusion protein preserves myocardial function after transient ischemia in mice" Circulation Feb. 7, 2012: 125(5):685-96. Doi: 10.1161/Circulation 111.070508.Epub Jan. 5, 2012.

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210040233A1 (en) * 2010-05-21 2021-02-11 Silver Creek Pharmaceuticals, Inc. Bi-Specific Fusion Proteins
US11673970B2 (en) * 2010-05-21 2023-06-13 Silver Creek Pharmaceuticals, Inc. Bi-specific fusion proteins
US11814443B2 (en) 2010-05-21 2023-11-14 Silver Creek Pharmaceuticals, Inc. Bi-specific fusion proteins
US11879002B2 (en) 2015-10-02 2024-01-23 Silver Creek Pharmaceuticals, Inc. Bi-specific therapeutic proteins, in vivo methods of use thereof and encoding nucleic acids thereof

Also Published As

Publication number Publication date
AU2011255238B2 (en) 2015-06-04
JP6822911B2 (en) 2021-01-27
CN103124788A (en) 2013-05-29
US20110286976A1 (en) 2011-11-24
US20160168269A1 (en) 2016-06-16
US20200048368A1 (en) 2020-02-13
US8691771B2 (en) 2014-04-08
JP2019196365A (en) 2019-11-14
ES2674567T3 (en) 2018-07-02
JP6174763B2 (en) 2017-08-02
US11814443B2 (en) 2023-11-14
JP2017225442A (en) 2017-12-28
US10407512B2 (en) 2019-09-10
US9238080B2 (en) 2016-01-19
JP2013530146A (en) 2013-07-25
BR112012029611A2 (en) 2017-07-25
CA2800173C (en) 2019-05-14
US20210214463A1 (en) 2021-07-15
WO2011146902A1 (en) 2011-11-24
JP2017000143A (en) 2017-01-05
US20210040233A1 (en) 2021-02-11
CN103124788B (en) 2016-01-13
US20140178380A1 (en) 2014-06-26
AU2011255238A1 (en) 2012-12-06
US11673970B2 (en) 2023-06-13
JP6200806B2 (en) 2017-09-20
US20170335015A1 (en) 2017-11-23
EP2571992A1 (en) 2013-03-27
US20230322951A1 (en) 2023-10-12
WO2011146902A9 (en) 2012-11-15
US20240101713A1 (en) 2024-03-28
EP2571992B1 (en) 2018-04-25
US20180237540A1 (en) 2018-08-23
US9982060B2 (en) 2018-05-29
CA2800173A1 (en) 2011-11-24
JP6965311B2 (en) 2021-11-10
US20110293579A1 (en) 2011-12-01
US9718892B2 (en) 2017-08-01
US10988547B2 (en) 2021-04-27

Similar Documents

Publication Publication Date Title
US11673970B2 (en) Bi-specific fusion proteins
US11879002B2 (en) Bi-specific therapeutic proteins, in vivo methods of use thereof and encoding nucleic acids thereof
CN105524176B (en) Bispecific fusion proteins
AU2013202341A1 (en) Bi-specific fusion proteins

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: MERRIMACK PHARMACEUTICALS, INC, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:NIELSEN, ULRIK;WICKHAM, THOMAS;SCHOEBERL, BIRGIT;AND OTHERS;SIGNING DATES FROM 20130321 TO 20130515;REEL/FRAME:046903/0155

AS Assignment

Owner name: SILVER CREEK PHARMACEUTICALS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MERRIMACK PHARMACEUTICALS, INC.;REEL/FRAME:049112/0597

Effective date: 20190401

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STCF Information on status: patent grant

Free format text: PATENTED CASE